[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

CN112313235A - Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases - Google Patents

Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases Download PDF

Info

Publication number
CN112313235A
CN112313235A CN201980039582.5A CN201980039582A CN112313235A CN 112313235 A CN112313235 A CN 112313235A CN 201980039582 A CN201980039582 A CN 201980039582A CN 112313235 A CN112313235 A CN 112313235A
Authority
CN
China
Prior art keywords
absent
substituted
phenyl
compound
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201980039582.5A
Other languages
Chinese (zh)
Inventor
南基然
金在昇
田義珍
兪東勳
徐武永
朴桐湜
J.艾克霍夫
G·齐施因斯基
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lead Discovery Center GmbH
Qurient Co Ltd
Original Assignee
Lead Discovery Center GmbH
Qurient Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lead Discovery Center GmbH, Qurient Co Ltd filed Critical Lead Discovery Center GmbH
Publication of CN112313235A publication Critical patent/CN112313235A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Communicable Diseases (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention relates to pyrazolo [1,5-a ] [1,3,5] triazine and pyrazolo [1,5-a ] pyrimidine derivatives and/or pharmaceutically acceptable salts thereof, the use of these derivatives as pharmaceutically active agents, in particular for the prevention and/or treatment of cell proliferative disorders, inflammatory disorders, immune disorders, cardiovascular disorders and infectious diseases. Furthermore, the present invention relates to a pharmaceutical composition containing at least one of the pyrazolo [1,5-a ] [1,3,5] triazine and pyrazolo [1,5-a ] pyrimidine derivatives and/or pharmaceutically acceptable salts thereof.

Description

Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases
Technical Field
The present invention relates to pyrazolo [1,5-a ] [1,3,5] triazine and pyrazolo [1,5-a ] pyrimidine derivatives and/or pharmaceutically acceptable salts thereof, the use of these derivatives as pharmaceutically active agents, in particular for the prevention and/or treatment of cell proliferative disorders, inflammatory disorders, immunological disorders, cardiovascular disorders and infectious diseases. Furthermore, the present invention relates to a pharmaceutical composition containing at least one of the pyrazolo [1,5-a ] [1,3,5] triazine and pyrazolo [1,5-a ] pyrimidine derivatives and/or pharmaceutically acceptable salts thereof.
Background
Cyclin-dependent kinase (CDK) family members that trigger passage through the cell cycle are considered attractive therapeutic targets, particularly for cancer. CDK family members that control other processes such as transcription and RNA processing have attracted little attention to date despite the emerging experimental evidence that they are involved in different pathological processes. Along with cell cycle control, CDK/cyclin complexes have also been identified as conserved components of the RNA polymerase II (pol II) transcription machinery (Bregman et al, 2000, Front biosci.5: 244-257). There are currently 20 known mammalian CDKs. Although CDK 7-13 has been linked transcriptionally, only CDK1, 2, 4 and 6 showed verifiable associations with the cell cycle. Among the mammalian CDKs, only CDK7 has robust kinase activity, regulating both cell cycle progression and transcription (Desai et al, 1995, mol.cell biol.15, 345-350).
The universal transcription factor TFIIH purified from mammalian cells is composed of 10 subunits, 7 of which (p62, p52, p44, p34, XPD, XPB and TTDA) form the core complex. The 3 subunits (cyclins H, MAT1 and CDK7) form a CDK Activated Kinase (CAK) which is linked to the core of TFIIH via the XPD (ATP-dependent helicase) subunit of the complex. During transcription initiation, the helicase activity of TFIIH opens the core promoter DNA, while CDK7 phosphorylates the serine 5 and 7 (Akhtar et al, 2009, mol.cell 34,387-393) of the C-terminal domain (CTD) of Pol II and other transcription factors that control the initiation to elongation transition (Larochelle et al, 2012, nat. strut. mol.biol.19, 1108-1115). CDK7 is therefore an essential factor in the transcriptional process, suggesting that CDK7 is a target for cancer therapy, in particular transcription dependent cancers.
CDK7 has long been asserted to have an essential role in cell metabolism and viability. Transcriptional CDK inhibitors down-regulate a number of short-lived anti-apoptotic proteins, such as the anti-apoptotic proteins myeloid cell leukemia protein-1 (Mcl-1), B-cell lymphoma ultralong protein (Bcl-xL) and XIAP (X-linked IAP), D-cyclin, c-Myc, Mdm-2 (leading to p53 stabilization), p21 whose transcription is mediated by nuclear factor-. kappa.B (NF-kB) and hypoxia-induced VEGFwaf1Protein (Shapiro GI.2006, J Clin Oncol; 24(11): 1770-83). Transcriptional non-selective cyclin-dependent kinase inhibitor flavopiridol through MTranscriptional repression and downregulation of cl-1 induces apoptosis in multiple myeloma cells. These findings support the previous hypothesis that CDK7 may be a valuable target for therapeutic drugs against malignancies and cell Cycle-related diseases (Lolli G and Johnson LN.2005.cell Cycle 4: 572-577).
The function of CDK7 as a regulator of overall transcription and CDK7 as a therapeutic target for the treatment of a number of diseases and syndromes is associated with mutations in regulatory regions that neutralize transcription factors, cofactors, chromatin regulators, and non-coding RNAs. These mutations can lead to cancer, autoimmune diseases, neurological disorders, developmental syndromes, diabetes, cardiovascular disease and obesity, among others. Some transcription factors control the suspended release and extension of RNA polymerase II and, when their expression or function is altered, can produce invasive tumor cells (c-Myc) or some form of Autoimmunity (AIRE) (Tong Ihn Lee and Richard A. Young, Cell,2013,152: 1237-1251). Thus, inhibition of the kinase activity of human CDK7 may inhibit certain transcription factors associated with oncogenes through inhibition of the overall transcriptional process, resulting in antiproliferative activity through function in cell cycle progression and transcriptional regulation. More importantly, CDK7 has been shown to regulate the exponential expression of oncogenic transcription factors more strongly than other housekeeping genes in cancer cells. Thus, inhibition of CDK7 may differentially affect the transcription of certain oncogenes and housekeeping genes, thus ensuring a therapeutic window. Thus, transcriptional regulation and pharmacological inhibition by the appropriate global transcriptional repression caused by CDK7 may be useful in the treatment of proliferative disorders, including cancer. As a global regulator of transcription, CDK7 is a therapeutic target for the treatment of diseases such as inflammation, viral replication such as HIV, EBV, cancer and cardiac hypertrophy.
HIV-1 gene expression is regulated by the viral transactivator protein (Tat), which induces the transcriptional elongation of HIV-1 long tandem repeats. This induction requires hyperphosphorylation of the C-terminal domain repeat of RNA polymerase II. To achieve this, Tat stimulates the CTD kinase associated with the universal transcription factor of the promoter complex, in particular CDK7 associated with TFIIH (Nekhai et al, Biochem J. (2002)364, 649-657). The inventors of US 615968 also described that Tat binds to CDK7 and this interaction increased the ability of CAK to phosphorylate CTD. The authors of US 615968 further disclosed that transcriptional activation of Tat is dependent on CDK7 kinase activity. In addition, Young Kyeung Kim and colleagues concluded that TFIIH recruitment and activation represent the rate-limiting step in the emergence of HIV from latency (Young Kyeung Kim, EMBO (2006)25, 3596-.
During human cytomegalovirus infection, the levels of CDK7 and CDK9, as well as the other components of the kinase complex MAT-1/cyclin H, were up-regulated. In addition, there is an increase in the kinase activity of CDK7 and CDK9 (Tamrakar et al, Journal of Virology,2005, 79; 15477-.
Many antiviral drugs target viral proteins. They have the disadvantage that viruses often develop resistance to these drugs. Antiviral drugs that target cellular proteins essential for viral processes, such as CDK7, may circumvent these drawbacks. These drugs may also be effective in treating several unrelated viruses, and their effects will be additive to traditional antiviral agents. CDK7 inhibitors that have the dual function of CDK activating kinases and transcriptional regulation are very effective in the treatment of several viruses.
Disclosure of Invention
It is an object of the present invention to provide compounds and/or pharmaceutically acceptable salts thereof useful as pharmaceutically active agents, particularly for the prevention and/or treatment of cell proliferative diseases, inflammatory diseases, immune diseases, cardiovascular diseases and infectious diseases, and compositions comprising at least one of these compounds and/or pharmaceutically acceptable salts thereof as a pharmaceutically active ingredient.
In one aspect, the present invention relates to pyrazolo-triazine or pyrazolo-pyrimidine compounds, which are defined by the general formula I
Figure BDA0002830360940000041
Wherein
X is independently at each occurrence selected from CH and N;
L1is absent or is independently selected at each occurrence from-NH-,-NH(CH2)-、-NH(C=O)-、-NHSO2-、-O-、-O(CH2) -, - (C ═ O) NH-, and- (C ═ O) (CH)2)-;
Q is independently at each occurrence selected from the group consisting of C3-C8 cycloalkyl, aryl, heteroaryl, heterocyclyl, and C1-C6 alkyl, wherein C1-C6 alkyl is-OR5、–N(R5)R5Aryl, heteroaryl and heterocyclyl, substituted with one or both,
C3-C8 cycloalkyl may be substituted by R3、R4And- (C ═ O) R5One or both of them may be substituted,
the heterocyclic radical may be substituted by R3、R4And- (C ═ O) R5One or both of them may be substituted,
the aryl OR heteroaryl group may be substituted by C1-C6 alkyl, -OR5、-N(R5)R5、-(C=O)R5One or both of halo, heteroaryl and heterocyclyl;
R1independently at each occurrence, selected from hydrogen and methyl;
R2independently at each occurrence, is selected from halogen, C1-C6 alkyl, C3-C10 cycloalkyl, -CN, - (C ═ O) CH3、-NR9R12And C1-C3 haloalkyl, any of which is optionally substituted;
R3independently at each occurrence, is selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R4independently at each occurrence, selected from hydrogen, halogen, -OR5、-N(R5)R5Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R5independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C3 haloalkyl, heteroaryl, heterocyclyl, substituted with halogen, -OR11、-N(R11)R11C1-C6 alkyl and optionally-OH or-NH2Heteroaryl substituted by one OR both of substituted C1-C6 alkyl, by halogen, -OR11、-N(R11)R11C1-C6 alkyl and optionally-OH or-NH2Heterocyclyl substituted with one or both of substituted C1-C6 alkyl;
z is any structure in the following group A;
Figure BDA0002830360940000051
group A
Wherein
X1Independently at each occurrence is selected from CR24And N;
X2independently at each occurrence is selected from CR25And N;
R6independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkyl substituted with-OH, C3-C10 cycloalkyl, C3-C10 heterocyclyl, - (C ═ O) NHR11、-NHR9、-NH(C=O)NHR11、-N(CH3)(C=O)CH3、-NH(C=O)R12、-NR9R12、-OR12And any structure in the following group B;
Figure BDA0002830360940000061
group B
R7Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, -NH (C ═ O) R12、-NR9R12、-OR12And any structure in group C below;
Figure BDA0002830360940000062
group C
R8And R10Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2、-OH、-OR5、-CN、-(C=O)R5、-(C=O)OR5、-(C=O)NH2、-(C=O)NHR21、-CH2(C=O)NHR21、-NH(C=O)R13、-NHS(=O)2R5、-S(=O)2NH2、-S(=O)2NHR21And by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R9independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OR5-CN, -C3-C10 cycloalkyl, -C3-C10 heterocyclyl and a substituted group-OH OR-OR5Substituted C1-C6 alkyl;
R11independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, and C3-C10 cycloalkyl;
R12absent OR independently at each occurrence selected from C1-C6 alkyl, substituted with-OR5or-N (R)5)R5Substituted C1-C6 alkyl, C6-C10 aryl, phenyl, benzyl, C3-C9 heteroaryl, C3-C6 heterocyclyl, benzyl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C9 heteroaryl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C6 heterocyclyl substituted with C1-C3 alkyl, and C3-C6 heterocyclyl substituted with 1 to 4 halogens and/or 1 to 4-NH (C ═ O) R13Substituted C6-C10 aryl;
R13independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl and C3-C10 cycloalkyl;
R14and R15Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, substituted with-OH or-NH2Substituted C1-C6 alkyl, C3-C10 cycloalkyl, - (C ═ O) R5,-(C=O)NHR21,-C(R9)(R11)OR21,-NH(C=O)R21,-NR9R21,-OR21,-OC(R9)(R11)(R21) C3-C10 heterocyclyl, by R4Substituted C3-C10 heterocyclyl, C3-C10 heteroaryl substituted by 1 to 4 halogens or C1-C3 alkyl, C6-C10 aryl such as phenyl and substituted by- (C ═ O) R5、-(C=O)OR5、-(C=O)NH2、-(C=O)NHR21、-CH2(C=O)NHR21、-NH(C=O)R13、-NHS(=O)2R5、-S(=O)2NH2or-S (═ O)2NHR21A substituted aryl group;
R16independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, - (C ═ O) R13And is-OR5Substituted C1-C6 alkyl;
R17、R18、R19and R20Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, C6-C10 aryl such as phenyl, -CN, -CHCF3NR9R11,-OH,-OR21,-NO2,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-NH(C=O)R13,-NHR5,-NHS(=O)2R5,-S(=O)2NH2,-S(=O)2NHR21And is substituted by-CN, -OH, -OR5、-(C=O)NHR5、-NH2、-NH(C=O)R5、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl;
R21independently at each occurrence, is selected from the group consisting of C1-C6 alkyl, C3-C10 cycloalkyl, C3-C10 heterocyclyl, C1-C3 haloalkyl, aryl, phenyl, benzyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl, aryl substituted by halogen or C1-C3 haloalkyl, C3-C10 heteroaryl substituted by 1 to 4 halogen or C1-C3 alkyl and R4Substituted C3-C10 heterocyclyl;
R22and R23Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OH, -OR5-CN and-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R24and R25Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2,-OH,-OR5,-CN,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-CH2(C=O)NHR21,-NH(C=O)R13,-NHS(=O)2R5,-S(=O)2NH2or-S (═ O)2NHR21And by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
provided that when Z is
Figure BDA0002830360940000081
When then R is6And R7One is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not CH3Cl or 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not phenyl, CH (CH)3)2、CH2CH3Or CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is F, R7Is H, R8Is H and R9Is H, then R10Is not F;
wherein,if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not F;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R4Is H, X is N, Z is phenyl, R6Is 1H-pyrazole, R7Is H, R8Is H, R9Is H and R10Is H, then R3Is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is H, R8Is H, R9Is H and R10Is H, then R7Is not Cl;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X isN, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Or enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates or pharmaceutically acceptable salts thereof of the above compounds.
In one embodiment, R1Is hydrogen and the compound has the general formula II
Figure BDA0002830360940000121
X, Q, L therein1、R2And Z is as defined above for formula II.
In one embodiment the invention relates to compounds having the general formula III
Figure BDA0002830360940000122
X, L therein1、R1、R2And Z is as defined above for formula I, and
Q1absent OR independently at each occurrence selected from aryl, heteroaryl, heterocyclyl, substituted with C1-C6 alkyl, -OR5、-N(R5)R5And aryl substituted by one OR both of halogen, C1-C6 alkyl, -OR5、-N(R5)R5And heteroaryl substituted with one or both of halogen and R29And R30Heterocyclic groups substituted with one or both of them;
R29absent OR independently at each occurrence selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R30independently at each occurrence, selected from hydrogen, halogen, -OR5、-N(R5)R5Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
wherein R is5、R9And R12As defined in claim 1;
L2absent or independently at each occurrence selected from-O-, -NH-, - (C ═ O) -, and- (C ═ O) NH-;
Y1independently at each occurrence, selected from CH, C (OH), and N;
Y2independently at each occurrence is selected fromCH、CR30O and N;
m is independently selected at each occurrence from 0, 1 and 2;
n is independently selected at each occurrence from 0 and 1;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not CH3Cl or 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not phenyl, CH (CH)3)2、CH2CH3Or CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is F, R7Is H, R8Is H and R9Is H, then R10Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is a group of atoms selected from the group consisting of CH,Y2is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is N, m is 1, N is 1, R30Is H, X is N, Z is phenyl, R6Is 1H-pyrazole, R7Is H, R8Is H, R9Is H and R10Is H, then R29Is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 2, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 2, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H andand R is10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 2, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is H, R8Is H, R9Is H and R10Is H, then R7Is not Cl;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2In the absence of the presence of the agent,Y1is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 2, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Or enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates or pharmaceutically acceptable salts thereof of the above compounds.
In one embodiment, the present invention relates to compounds having the general formula Ia
Figure BDA0002830360940000181
Wherein
X is independently at each occurrence selected from CH and N;
Y1independently at each occurrence, selected from CH, C (OH), and N;
Y2independently at each occurrence, selected from CH, CR4O and N;
m is independently selected at each occurrence from 0, 1 and 2;
n is independently selected at each occurrence from 0 and 1;
L1is absent or independently selected at each occurrence from-NH-, -NH (CH)2)-、-NH(C=O)-、-NHSO2-、-O-、-O(CH2) -, - (C ═ O) NH-, and- (C ═ O) (CH)2)-;
Q is absent or independently at each occurrence selected from the group consisting of heterocyclyl, C3-C6 heteroaryl, aryl such as phenyl, aryl substituted with halogen;
L2absent or independently at each occurrence selected from-O-, -NH-, - (C ═ O) -, and- (C ═ O) NH-;
R1independently at each occurrence, selected from hydrogen and methyl;
R2independently at each occurrence, is selected from halogen, C1-C6 alkyl, C3-C10 cycloalkyl, -CN, - (C ═ O) CH3、-NR9R12And C1-C3 haloalkyl, any of which is optionally substituted;
R3absent or independently at each occurrence selected from hydrogen, -OH, halogen, -NH2、-NR9R12、-NH(C=O)R5、-(C=O)NH2Heterocyclyl, C1-C6 alkyl and substituted by-OH or-NH2Substituted C1-C6 alkyl;
R4absent OR independently at each occurrence selected from hydrogen, halogen, -OH, -OR5、-NH2(═ O), C1-C6 alkyl, and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R5independently at each occurrence, is selected from the group consisting of C1-C6 alkyl, C3-C6 cycloalkyl, C3-C10 heterocyclyl, C1-C3 haloalkyl, and substituted with halogen, -OH, -NH2C1-C6 alkyl and optionally-OH or-NH2Substituted C1-C6 alkyl substituted C3-C10 heterocyclyl;
z is any structure in the following group A;
Figure BDA0002830360940000191
group A
Wherein
X1Independently at each occurrence is selected from CR24And N;
X2independently at each occurrence is selected from CR25And N;
R6independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C3-C10 cycloalkyl, C3-C10 heterocyclyl, - (C ═ O) NHR11、-NHR9、-NH(C=O)NHR11、-N(CH3)(C=O)CH3、-NH(C=O)R12、-NR9R12、-OR12And any structure in the following group B;
Figure BDA0002830360940000201
group B
R7Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, -NH (C ═ O) R12、-NR9R12、-OR12And any structure in group C below;
Figure BDA0002830360940000202
group C
R8And R10Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2,-OH,-OR5,-CN,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-CH2(C=O)NHR21,-NH(C=O)R13,-NHS(=O)2R5,-S(=O)2NH2,-S(=O)2NHR21And by OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R9independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OR5-CN, -C3-C10 cycloalkyl, -C3-C10 heterocyclyl and a substituted group-OH OR-OR5Substituted C1-C6 alkyl;
R11independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, and C3-C10 cycloalkyl;
R12absent OR independently at each occurrence selected from C1-C6 alkyl, substituted with-OR5or-N (R)5)R5Substituted C1-C6 alkyl, C6-C10 aryl, phenyl, benzyl, C3-C9 heteroaryl, C3-C6 heterocyclyl, benzyl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C9 heteroaryl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C6 heterocyclyl substituted with C1-C3 alkyl, and C3-C6 heterocyclyl substituted with 1 to 4 halogens and/or 1 to 4-NH (C ═ O) R13Substituted C6-C10 aryl;
R13independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl and C3-C10 cycloalkyl;
R14and R15Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, substituted with-OH or-NH2Substituted C1-C6 alkyl, C3-C10 cycloalkyl, - (C ═ O) R5,-(C=O)NHR21,-C(R9)(R11)OR21,-NH(C=O)R21,-NR9R21,-OR21,-OC(R9)(R11)(R21) C3-C10 heterocyclyl, by R4Substituted C3-C10 heterocyclyl, C3-C10 heteroaryl substituted by 1 to 4 halogens or C1-C3 alkyl, C6-C10 aryl such as phenyl and substituted by- (C ═ O) R5、-(C=O)OR5、-(C=O)NH2、-(C=O)NHR21、-CH2(C=O)NHR21、-NH(C=O)R13、-NHS(=O)2R5、-S(=O)2NH2or-S (═ O)2NHR21A substituted aryl group;
R16independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, - (C ═ O) R13And is-OR5Substituted C1-C6 alkyl;
R17、R18、R19and R20Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, C6-C10 aryl such as phenyl, -CN, -CHCF3NR9R11,-OH,-OR21,-NO2,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-NH(C=O)R13,-NHR5,-NHS(=O)2R5,-S(=O)2NH2,-S(=O)2NHR21And is substituted by-CN, -OH, -OR5、-(C=O)NHR5、-NH2、-NH(C=O)R5、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl;
R21independently at each occurrence, is selected from the group consisting of C1-C6 alkyl, C3-C10 cycloalkyl, C3-C10 heterocyclyl, C1-C3 haloalkyl, aryl, phenyl, benzyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl, aryl substituted by halogen or C1-C3 haloalkyl, C3-C10 heteroaryl substituted by 1 to 4 halogen or C1-C3 alkyl, and R4Substituted C3-C10 heterocyclyl;
R22and R23Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OH, -OR5-CN and-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R24and R25Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2,-OH,-OR5,-CN,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-CH2(C=O)NHR21,-NH(C=O)R13,-NHS(=O)2R5,-S(=O)2NH2or-S (═ O)2NHR21And by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
provided that when Z is
Figure BDA0002830360940000221
When then R is6And R7One is not H.
Wherein if R is1Is H, R2Is CH (CH)3)2,R3Is NH2,R4Is H, L1Absence, Q is absent, L2Absent, X is N, N is 1, Y1Is N, Y2Is CH, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not CH3Cl or 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not phenyl, CH (CH)3)2、CH2CH3Or CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R6Is F, R7Is H, R8Is H and R9Is H, then R10Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is CH3,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not F;
wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is CH3,R4Is H, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is- (C ═ O) -, Q is absent, L2Is absent, Y1Is N, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R7Is a compound of formula (I) wherein the compound is H,R8is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is- (C ═ O) -, Q is absent, L2Is absent, Y1Is N, Y2Is CH, n is 1, R3Is NH2,R4Is H, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 0, R3Is H, R4Is H, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 0, R3Is H, R4Is H, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 0, R3Is H, R4Is H, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,R3Is NH2,R4Is H, L1Is O, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is CH, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is H, R4Is H, L1Is NH, Q is absent, L2Absent, X is N, N is 0, Y1Is CH, Y2Is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (C)H3)2,R3Is NH2,R4Is H, L1Absence, Q is absent, L2Absent, X is N, N is 0, Y1Is N, Y2Is CH, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is NH2,R4Is H, L1Is NH, Q is absent, L2Absent, X is N, N is 1, Y1Is CH, Y2Is CH, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,R3Is H, R4Is NH2,L1Absence, Q is absent, L2Absent, X is N, N is 1, Y1Is N, Y2Is CH, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is CH3,R4Is H, Z is phenyl, R6Is H, R8Is H, R9Is H and R10Is H, then R7Is not Cl;
wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 1, R3Is NH2,R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is O, Q is absent, L2Is absent, Y1Is CH, Y2Is CH, n is 1, R3Is NH2,R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Absence, Q is absent, L2Is absent, Y1Is N, Y2Is CH, n is 1, R3Is NH2,R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is NH, Q is absent, L2Is absent, Y1Is CH, Y2Is N, N is 0, R3Is H, R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Absence, Q is absent, L2Is absent, Y1Is N, Y2Is CH, n is 0, R3Is NH2,R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is NH, Q is absent, L2Is absent, Y1Is CH, Y2Is CH, n is 1, R3Is NH2,R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is- (C ═ O) -, Q is absent, L2Is absent, Y1Is N, Y2Is N, N is 1, R3Is H, R4Is H, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Is- (C ═ O) -, Q is absent, L2Is absent, Y1Is N, Y2Is CH, n is 1, R3Is NH2,R4Is H, Z is a phenyl group,R6is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2X is N, L1Absence, Q is absent, L2Is absent, Y1Is N, Y2Is CH, n is 1, R3Is NH, R4Is NH2Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Or enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates or pharmaceutically acceptable salts thereof of the above compounds.
In one embodiment, the invention relates to compounds having the general formula IV
Figure BDA0002830360940000261
X, L therein1And R2As defined above for formula I;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined above for formula III;
wherein Z1Is any one of the following group D;
Figure BDA0002830360940000271
group D
Wherein R is6、R7、R8、R9And R10As defined above for formula I.
In one embodiment, the present invention relates to compounds having the general formula V
Figure BDA0002830360940000272
X, X therein1、X2、L1、R2、R6、R22And R23As defined above for formula I;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined above for formula III.
In one embodiment, the present invention relates to compounds having the general formula VI
Figure BDA0002830360940000273
X, L therein1And R2As defined above for formula I;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined above for formula III;
X3independently at each occurrence is selected from CR10And N;
R26、R27and R28Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OR5-CN and-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R5、R9and R10As defined above for formula I;
R7is any one of the following group E;
Figure BDA0002830360940000281
group E
Wherein R is8And R14–R20As described above for formula IAnd (4) defining.
In one embodiment, the invention relates to compounds having the general formula VII
Figure BDA0002830360940000282
X, L therein1、R2、R6、R22、R23、R24And R25As defined above for formula I;
m、n、Y1、Y2、L2、R29、R30and Q1As defined above for formula III.
In one embodiment, the present invention relates to compounds having the general formula VIII
Figure BDA0002830360940000291
X, L therein1、R2、R6And R10As defined above for formula I.
In one embodiment, the invention relates to compounds having the general formula IX
Figure BDA0002830360940000292
Wherein L is1、R2、R6、R22、R23、R24And R25As defined above for formula I, and
X3as defined above for formula VI;
wherein Q2Is any one of the following group F;
Figure BDA0002830360940000293
group F
R31And R32Absent OR independently at each occurrence selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
wherein R is5、R9And R12As defined above for formula I.
In one embodiment, the invention also relates to pharmaceutically acceptable salts of the compounds according to the invention as defined herein.
In one embodiment, the compound according to the present invention is a compound selected from structures 1-279 further listed in table 11 below.
In another aspect, the present invention also relates to a pharmaceutical composition comprising as active ingredient a compound according to the invention as defined herein and at least one pharmaceutically acceptable carrier, excipient and/or diluent.
In one aspect, the invention also relates to a compound according to the invention as defined herein for use as a medicament or pharmaceutically active agent, wherein said medicament or pharmaceutically active agent preferably has inhibitory activity on cyclin dependent kinase 7(CDK 7).
In one aspect, the invention also relates to a compound according to the invention as defined herein for use in a method for the prevention and/or treatment of a disease associated with inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or several Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), wherein said disease is selected from the group consisting of a proliferative disease, an infectious disease including an opportunistic disease, an immune disease, an autoimmune disease and an inflammatory disease.
In one embodiment, the disease associated with inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or more Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), is a disease associated with, accompanied by, caused by and/or induced by CDK7 dysfunction and/or hyperactivity. In one embodiment, the disease associated with inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or more Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), is a proliferative disease. In one embodiment, the proliferative disease is cancer.
In one embodiment, the cancer is selected from the group consisting of adenocarcinoma, choroidal melanoma, acute leukemia, acoustic neuroma, ampulla cancer, anal cancer, astrocytoma, basal cell carcinoma, pancreatic cancer, desmoid tumor, bladder cancer, bronchial cancer, estrogen-dependent and independent breast cancer, burkitt lymphoma, uterine cancer, cancer of the uterine body, cancer of unknown primary tumor (CUP syndrome), colorectal cancer, cancer of the small intestine, tumor of the small intestine, ovarian cancer, endometrial cancer, ependymoma, types of epithelial cancer, ewing's tumor, gastrointestinal tumor, gastric cancer, cancer of the gallbladder epithelium, cancer of the uterus, cancer of the cervix, glioblastoma, gynecological tumor, otorhinolaryngological tumor, hematological tumor, hairy cell leukemia, cancer of the urethra, skin cancer, skin testicular cancer, brain tumor (glioma), brain metastasis tumor, testicular cancer, pituitary tumor, Carcinoid tumor, Kaposi's sarcoma, laryngeal cancer, germ cell tumor, bone cancer, colorectal cancer, head and neck tumor (tumor of ear, nose and throat region), colon cancer, craniopharyngeal tumor, oral cavity cancer (cancer of oral region and lip), cancer of central nervous system, liver cancer, liver metastasis tumor, leukemia, eyelid tumor, lung cancer, lymphoma, stomach cancer, malignant melanoma, malignant neoplastic change, gastrointestinal malignant tumor, breast cancer, rectal cancer, medulloblastoma, melanoma, meningioma, Hodgkin/non-Hodgkin lymphoma, mycosis fungoides, nasal cancer, schwannoma, neuroblastoma, kidney cancer, renal cell carcinoma, oligodendroglioma, esophageal cancer, osteolytic and osteogenic cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, plasmacytoma, prostate cancer, pharyngeal cancer, rectal cancer, retinoblastoma, prostate cancer, cervical cancer, renal cancer, neuroblastoma, melanoma, neuroblastoma, Vaginal cancer, thyroid cancer, esophageal cancer, T-cell lymphoma, thymoma, ductal cancer, ocular tumor, urinary tract cancer, urinary tract tumor, urothelial cancer, vulva cancer, wart appearance, soft tissue tumor, soft tissue sarcoma, wilms' tumor, cervical cancer, tongue cancer, invasive ductal cancer, invasive lobular cancer, ductal cancer in situ, lobular cancer in situ, small cell lung cancer, non-small cell lung cancer, bronchial adenoma, pleuropneumonias, mesothelioma, brain stem glioma, hypothalamic glioma, cerebellar astrocytoma, brain astrocytoma, neuroectodermal tumor, pineal tumor, uterine sarcoma, salivary gland carcinoma, anal gland adenocarcinoma, mast cell tumor, pelvic tumor, ureteral tumor, hereditary renal carcinoma, papillary renal carcinoma in eye, melanoma, hepatocellular carcinoma, bile duct cancer, mixed hepatocyte cholangiocarcinoma, hepatoma carcinoma, Squamous cell carcinoma, malignant melanoma, merkel cell skin cancer, non-melanoma skin cancer, hypopharynx cancer, nasopharyngeal cancer, oropharyngeal cancer, oral cancer, squamous cell cancer, oral melanoma, AIDS-related lymphoma, cutaneous T-cell lymphoma, central nervous system lymphoma, malignant fibrous histiocytoma, lymphosarcoma, rhabdomyosarcoma, malignant histiocytosis, fibrosarcoma, angiosarcoma, hemangiothecoma, Leiomyosarcoma (LMS), canine breast cancer, and feline breast cancer.
In one embodiment, the infectious disease, including an opportunistic disease, is selected from the group consisting of AIDS, adenovirus infection, alveolar echinococcosis (AHD), amebiasis, angiostrongylosis, heterojulopathy, anthrax, babesiosis, venosasis, ascariasis, schistosomiasis (schistosomiasis), human blastocyst protozoa infection, Lyme borreliosis, botulism, Breynard diarrhea, Brucella disease, Bovine Spongiform Encephalopathy (BSE), candidiasis, capillariosis, Chronic Fatigue Syndrome (CFS), Chagas, chicken pox, Chlamydia pneumoniae infection, cholera, chronic fatigue syndrome, Creutzfeldt-Jakob disease (CJD), cremopsis, cutaneous larval immigration disease (CLM), coccidioidomycosis, conjunctivitis, Coxsackie virus A16(Cox A16), cryptococcosis, cryptosporidiosis, West Nile fever, coccidiosis, Cerebral cysticercosis, cytomegalovirus infection, dengue fever, canine polypore tapeworm infection, Ebola Hemorrhagic Fever (EHF), Alveolar Echinococcosis (AE), encephalitis, intracolonic amoeba infection, dispar amoeba infection, hamaunaeba infection, bordetella infection, enterobiasis infection, enterovirus infection (polio/non-polio), Epstein Barr virus infection, escherichia coli infection, food-borne infection, foot and mouth disease, fungal dermatitis, fungal infection, gastroenteritis, group a streptococcal disease, group B streptococcal disease, hansenese disease (leprosy), hantavirus pulmonary syndrome, head lice (lice), helicobacter pylori infection, blood disease, hendra virus infection, hepatitis (HCV, HBV), herpes zoster (shingles), HIV infection, human ehrlichiosis, human parainfluenza virus infection, herpes zoster infection, and other infections, Influenza, isosporadic disease, lassa fever, leishmaniasis, Visceral Leishmaniasis (VL), malaria, marburg hemorrhagic fever, measles, meningitis, Mycobacterium Avium Complex (MAC) infection, naesvarella infection, nosocomial infection, nonpathogenic intestinal amoeba infection, onchocerciasis, posttesticular trematosis, papilloma virus infection, parvovirus infection, plague, pneumocystis pneumonia (PCP), polyoma virus infection, Q fever, rabies, Respiratory Syncytial Virus (RSV) infection, rheumatic fever, rift valley fever, rotavirus infection, ascaris infection, salmonellosis, scabies, shigellosis, herpes zoster, narcolepsy, streptococcal infection, taenia infection, tetanus, toxic shock syndrome, tuberculosis, duodenum, vibrio parahaemolyticus infection, vibrio septicemia, viral hemorrhagic fever, meningitis, herpes, narcolepsy, herpes zoster, herpes zoster, Warts, water-borne infectious diseases, varicella-zoster virus infection, pertussis and yellow fever.
In one embodiment, the immune and/or autoimmune disease is selected from the group consisting of asthma, diabetes, rheumatic diseases, rejection of transplanted organs and tissues, rhinitis, chronic obstructive pulmonary disease, osteoporosis, ulcerative colitis, sinusitis, lupus erythematosus, recurrent infections, atopic dermatitis/eczema and occupational allergies, food allergies, drug allergies, severe allergic reactions, allergies, manifestations of allergic disease, primary immunodeficiency, antibody deficient states, cell-mediated immunodeficiency, severe combined immunodeficiency, DiGeorge syndrome, hyper IgE syndrome (HIES), Wiskott-Aldrich syndrome (WAS), ataxia telangiectasia, immune-mediated cancer, leukocyte deficiency, autoimmune diseases, Systemic Lupus Erythematosus (SLE), Rheumatoid Arthritis (RA), Multiple Sclerosis (MS), and autoimmune diseases, Immune-mediated or type 1 diabetes, immune-mediated glomerulonephritis, scleroderma, pernicious anemia, alopecia, pemphigus vulgaris, myasthenia gravis, inflammatory bowel disease, crohn's disease, psoriasis, autoimmune thyroid disease, hashimoto's disease, dermatomyositis, goodpasture's syndrome (GPS), Myasthenia Gravis (MG), sympathetic ophthalmia, lens uveitis, chronic invasive hepatitis, primary biliary cirrhosis, autoimmune hemolytic anemia, and weichhoff's disease.
In one embodiment, the inflammatory disease is caused, induced, caused and/or enhanced by a bacterium, virus, prion, parasite, fungus, and/or caused by a stimulatory, traumatic, metabolic, allergic, autoimmune or idiopathic agent.
In one embodiment, the inflammatory disease is selected from inflammatory diseases of the Central Nervous System (CNS), inflammatory rheumatic diseases, inflammatory diseases of the blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, uveitis inflammatory diseases and inflammatory diseases of the throat.
In one embodiment, the inflammatory disease is selected from inflammatory diseases of the Central Nervous System (CNS), inflammatory rheumatic diseases, inflammatory diseases of the blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, uveitis, inflammatory diseases of the throat, wherein preferably the inflammatory disease is selected from abscesses, acanthamoeba infection, acne vulgaris, mycosis, acute inflammatory skin diseases, acute laryngeal infection in adults, acute multifocal squamous pigment epithelium lesion, acute (thermal) injury, acute retinal necrosis, acute purulent otitis media, algal disorders, allergic contact dermatitis, amyloidosis vascular edema, ankylosing spondylitis, aspergillosis, atopic dermatitis, pseudorabies, autoantibodies in vasculitis, bacterial disorders, bacterial inflammation, bacterial meningitis, Behcet's Disease (BD), Mariothis gallinarum disease, Creutzfeldt-Jakob disease, Borna disease, Brucella disease, bullous myringitis, bursitis, candidiasis, Canine fever encephalomyelitis, canine hemorrhagic fever, Canine herpes virus encephalomyelitis, cholesteatoma, Chronic Granulomatosis (CGD), Chronic inflammatory skin disease, Chronic relapsing encephalomyelitis, Chronic pyogenic otitis media, Ocular Cicatricial Pemphigoid (OCP), common upper respiratory tract infections, granuloma, Crohn's disease, cryptococcosis, dermatomyositis, diphtheria, Discoid Lupus Erythematosus (DLE), drug-induced vasculitis, drug or hypersensitivity reactions, encephalopathy, eosinophilic meningoencephalitis, Erythema Multiforme (EM), feline leukemia virus, feline immunodeficiency virus, peritonitis, feline polio virus encephalitis, feline spongiform encephalopathy, Fibromyalgia, Fuchs heterochrosis uveitis, gastroesophageal (laryngopharyngeal) reflux disease, giant cell arteritis, melioidosis, glaucomatous cyclitis syndrome, gonorrheal granulomatosis, Granulomatous Meningoencephalitis (GME), herpes simplex, histoplasmosis, idiopathic diseases, idiopathic inflammatory disorders, immune and idiopathic disorders, infection of an immunocompromised host, infectious canine hepatitis, inhalation laryngitis, interstitial nephritis, irritant contact dermatitis, juvenile rheumatoid arthritis, kawasaki disease, Lakrauss virus encephalitis, laryngeal abscess, laryngotracheobronchitis, leishmaniasis, lens-induced uveitis, leprosy disease, leptospirosis, leukemia, lichen planus, lupus, lymphoma, meningitis, meningoencephalitis in dogs, miscellaneous meningitis/meninges, microscopic polyangiitis, polymyositis, meningitis, and meningitis, Multifocal choroiditis, multifocal distemper encephalomyelitis in mature animals, multiple sclerosis, myotonic-phonic disorder (MTD), fungal disease of the CNS, necrotizing encephalitis, neosporosis, old dog encephalitis, onchocerciasis, parasitic encephalomyelitis, parasitic infection, pars plana, parvoviral encephalitis, pediatric laryngitis, pollution and inhalation hypersensitivity, polymyositis, canine distemper heat encephalitis after vaccination, prion protein-induced disease, protozoonosis, protozoan encephalitis-encephalomyelitis, psoriasis, psoriatic arthritis, haba dog encephalitis, radiation injury, radiation laryngitis, radiation necrosis, recurrent polychondritis, Reiter's syndrome, retinitis pigmentosa, retinoblastoma, rheumatoid arthritis, rickettsialism, rocky mountain spotted fever, salmon mesovirus (SPD), Sarcoidosis, schistosomiasis, scleroderma, rhinoscleroderma, creeping choroiditis, canine tremor, sjogren's syndrome, spastic laryngitis, spirochaetes (syphilis) disease, spongiform dermatitis, sporotrichosis, steroid responsive meningitis-arteritis, Stevens-Johnson syndrome (SJS, severe EM), epiglottitis, sympathetic ophthalmia, heterodera, syphilis, systemic vasculitis in sarcoidosis, takayasu's arteritis, tendonitis (tenosynovitis), thromboangiitis obliterans (berger disease), tick-borne encephalitis in dogs, Toxic Epidermal Necrolysis (TEN), toxocariasis, toxoplasmosis, trauma, traumatic laryngitis, trichinosis, trypanosomiasis, tuberculosis, tularemia, ulcerative colitis, urticaria (urticaria), vasculitis, and malignant tumors, vasculitis, and rheumatoid arthritis, Vasculitis in idiopathic inflammatory myopathies, vasculitis of the central nervous system, vasculitis secondary to bacterial, fungal and parasitic infections, viral disorders, viral laryngitis, vitiligo, vocal cord abuse, vocal cord hemorrhage, Vogt-Koyanagi-Harada syndrome (VKH), Wegener's granulomatosis, and Whipple's disease.
The present invention also relates to a method for the treatment and/or prevention of a disease associated with inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or several Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), wherein said disease is selected from the group consisting of a proliferative disease, an infectious disease including an opportunistic disease, an immune disease, an autoimmune disease and an inflammatory disease, wherein said method for treatment and/or prevention comprises administering a compound according to the invention as defined herein to a patient in need thereof.
In one embodiment, the patient in need thereof is a mammal. In one embodiment, the patient in need thereof is a human. In another embodiment, the patient in need thereof is a non-human animal.
In one embodiment, the disease prevented or treated in the method is as defined herein.
The present invention also relates to the use of a compound according to the invention as defined herein for the manufacture of a medicament for the prevention and/or treatment of a disease associated with inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or several Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), wherein the disease is selected from the group consisting of a proliferative disease, an infectious disease including an opportunistic disease, an immune disease, an autoimmune disease and an inflammatory disease as defined herein.
Further advantageous features, aspects and details of the invention will become apparent from the dependent claims, the description, the embodiments and the drawings.
The compounds of the invention are highly potent inhibitors of CDK7 threonine/serine kinase and/or its complex CDK7/MAT 1/CycH. The compounds of the invention are suitable for use as pharmaceutically active agents. The compounds of the present invention are useful in the treatment of disorders associated with, accompanied by, caused by and/or induced by CDK7 and its complexes, in particular hyperactivity or dysfunction thereof. Accordingly, the compounds of the present invention are useful in the treatment of CDK7 related diseases or disorders and CDK7 complex-induced disorders.
The compounds of the present invention may also be used in the manufacture of a medicament or pharmaceutical composition for the treatment of disorders associated with, accompanied, caused by and/or induced by CDK7 and its complexes, in particular hyperactivity or dysfunction thereof. The compounds of the present invention are further used in the manufacture of a medicament or pharmaceutical composition for the treatment and/or prevention of CDK7 and its complex-induced disorders.
The term "optionally substituted" as used herein means that a hydrogen atom present and attached to a member atom within a group or that several such hydrogen atoms may be replaced by suitable groups such as halogens including fluorine, halogen,C1-C3Alkyl radical, C1-C3Haloalkyl, methylhydroxy, COOMe, C (O) H, COOH, OMe or OCF3Instead.
The term "alkyl" refers to a monovalent straight, branched, or cyclic chain saturated aliphatic hydrocarbon radical having a number of carbon atoms within the specified range. Thus, for example, "C1-C6Alkyl "refers to the hexyl and pentyl isomers and any of n-, iso-, sec-and tert-butyl, n-and iso-propyl, cyclopropyl, ethyl and methyl.
The term "alkenyl" refers to a monovalent straight or branched chain aliphatic hydrocarbon radical containing one carbon-carbon double bond and having a number of carbon atoms within the specified range. Thus, for example, "C2-C6Alkenyl "means all hexenyl and propenyl isomers as well as 1-butenyl, 2-butenyl, 3-butenyl, isobutenyl, 1-propenyl, 2-propenyl, and ethenyl.
The term "cycloalkyl", alone or in combination with any other term, means a group having 3 to 8 carbon atoms, such as an optionally substituted or unsubstituted cyclic hydrocarbon group, unless otherwise defined. Thus, for example, "C3-C8Cycloalkyl "refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
The term "haloalkyl" refers to an alkyl group, as defined herein, substituted with at least one halogen. Examples of straight or branched chain "haloalkyl" useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, and t-butyl independently substituted with one or more halogens. The term "haloalkyl" should be construed to include groups such as-CHF2、–CF3、-CH2-CH2-F、-CH2-CF3And the like.
The term "heteroalkyl" refers to an alkyl group in which one or more carbon atoms have been replaced with a heteroatom such as O, N or S. For example, if the carbon atom of the alkyl group attached to the parent molecule is replaced with a heteroatom (e.g., O, N or S), the resulting heteroalkyl group is each an alkoxy group (e.g., -OCH)3Etc.), amines (e.g., -NHCH)3、-N(CH3)2Etc.) or thioalkyl (e.g., -SCH)3Etc.). If the non-terminal carbon atom of the alkyl group not attached to the parent molecule is replaced with a heteroatom (e.g., O, N or S), the resulting heteroalkyl group is, respectively, an alkyl ether (e.g., -CH)2CH2-O-CH3Etc.), alkylamines (e.g., -CH)2NHCH3、-CH2N(CH3)2Etc.) or thioalkyl ethers (e.g., -CH2-S-CH3)。
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "phenyl" as used herein means an optionally substituted or unsubstituted phenyl group.
As used herein, the term "benzyl" means an optionally substituted or unsubstituted benzyl.
The term "heteroaryl" refers to (i) optionally substituted 5-and 6-membered heteroaromatic rings and (ii) optionally substituted 9-and 10-membered bicyclic fused ring systems wherein at least one ring is aromatic, wherein the heteroaromatic ring or bicyclic fused ring system contains 1 to 4 heteroatoms independently selected from N, O and S, wherein each N is optionally in the form of an oxide, and each N in a ring that is not aromatic is optionally S (o) or S (o)2. Suitable 5-and 6-membered heteroaromatic rings include, for example, pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thienyl, furanyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, and thiadiazolyl. Suitable 9-and 10-membered heterobicyclic fused ring systems include, for example, benzofuranyl, indolyl, indazolyl, naphthyridinyl, isobenzofuranyl, benzopiperidinyl, benzisoxazolyl, benzoxazolyl, chromenyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, isoindolyl, benzodioxolyl, benzofuranyl, imidazo [1,2-a ]]Pyridyl, benzotriazolyl, indolinyl, dihydroisoindolyl, indazolyl, indolinyl, isoindolinyl, quinoxalinyl, quinazolinyl, 2, 3-dihydrobenzofuranyl and 2, 3-dihydrobenzo-1, 4-diylAn oxacyclopentenyl group.
The term "heterocyclyl" refers to (i) an optionally substituted 4 to 8 membered saturated and unsaturated but non-aromatic monocyclic ring containing at least one carbon atom and 1 to 4 heteroatoms, (ii) an optionally substituted bicyclic ring system containing 1 to 6 heteroatoms, and (iii) an optionally substituted tricyclic ring system, wherein each ring of (ii) or (iii) is independently fused or bridged to one or more other rings, and each ring is saturated or unsaturated but non-aromatic, and wherein each heteroatom of (i), (ii), and (iii) is independently selected from N, O and S, wherein each N is optionally in the form of an oxide, and each S is optionally oxidized to (S) or S (o)2. Suitable 4-to 8-membered saturated heterocyclic groups include, for example, azetidinyl, piperidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, pyrrolidinyl, imidazolidinyl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl, hexahydropyrimidinyl, thiomorpholinyl, thiaazepanyl, azepanyl, diazepanyl, tetrahydropyranyl, tetrahydrothiopyranyl, dioxanyl, and azocyclooctyl. Suitable unsaturated heterocycles include those corresponding to the saturated heterocycles listed in the preceding sentence in which the single bond is replaced by a double bond. It should be understood that the particular rings and ring systems suitable for use in the present invention are not limited to those listed herein and in the preceding paragraphs. These rings and ring systems are merely representative.
Pharmaceutically acceptable salts
Examples of pharmaceutically acceptable addition salts include, but are not limited to, non-toxic inorganic and organic acid addition salts, such as acetate derived from acetic acid, aconitate derived from aconitic acid, ascorbate derived from ascorbic acid, benzenesulfonate derived from benzenesulfonic acid, benzoate derived from benzoic acid, cinnamate derived from cinnamic acid, citrate derived from citric acid, pamoic acid salt derived from pamoic acid, heptanoate derived from heptanoic acid, formate derived from formic acid, fumarate derived from fumaric acid, glutamate derived from glutamic acid, glycolate derived from glycolic acid, hydrochloride derived from hydrochloric acid, hydrobromide derived from hydrobromic acid, lactate derived from lactic acid, maleate derived from maleic acid, malonate derived from malonic acid, mandelate derived from mandelic acid, methanesulfonate derived from methanesulfonic acid, hydrobromide derived from hydrobromic acid, lactate derived from lactic acid, maleate derived from maleic acid, malonate derived from malonic acid, mandelate derived from mandelic acid, mandelate derived from methanesulfonic acid, mesylate derived from methanesulfonic acid, Naphthalene-2-sulfonate derived from naphthalene-2-sulfonic acid, nitrate derived from nitric acid, perchlorate derived from perchloric acid, phosphate derived from phosphoric acid, phthalate derived from phthalic acid, salicylate derived from salicylic acid, sorbate derived from sorbic acid, stearate derived from stearic acid, succinate derived from succinic acid, sulfate derived from sulfuric acid, tartrate derived from tartaric acid, p-toluenesulfonic acid derived from p-toluenesulfonic acid, and the like. These salts may be formed by procedures well known and described in the art.
Other acids which may not be considered pharmaceutically acceptable, such as oxalic acid, may be used to prepare salts useful as intermediates in obtaining the chemical compounds of the invention and their pharmaceutically acceptable acid addition salts.
In another embodiment, the compounds of the invention are used according to the invention in their respective free base forms.
The metal salts of the chemical compounds of the present invention include alkali metal salts, such as sodium salts of the carboxylic group-containing chemical compounds of the present invention.
The chemical compounds of the present invention may be provided in unsolvated or solvated forms along with pharmaceutically acceptable solvents such as water, ethanol, and the like. Solvated forms may also include hydrated forms such as monohydrate, dihydrate, hemihydrate, trihydrate, tetrahydrate and the like. In general, the solvated forms are considered equivalent to unsolvated forms for the purposes of the present invention.
Other aspects of the invention are illustrated and exemplified by the following schemes, examples, tables, and program descriptions, which are provided for purposes of illustration only and not for limitation of the invention. The scope of the invention is limited only by the appended claims.
Tables and drawings
Reference will now be made to the table in which
Table 1 shows the results for selected compounds of the inventionActivity data in CDK1, CDK2, CDK5 and CDK7 enzyme assays. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 100 nM; b ═ IC50Greater than 100nM but less than 1,000 nM; c ═ IC50Greater than 1,000 nM. Table 1 also shows selectivity data in CDK1/CDK7, CDK2/CDK7 and CDK5/CDK7 for selected compounds of the invention. Selectivities are indicated as CDK1/CDK7, CDK2/CDK7 and CDK5/CDK7, and the following legends are used: a is higher than 200 times; b is less than 200 times but more than 20 times; c is less than 20 times.
Table 2 shows the activity data of the cellular HCT116 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 3 shows the activity data of the cellular H460 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 4 shows the activity data of the cellular mm.1s viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 5 shows the activity data of the cellular MV4-11 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 6 shows the activity data of the cellular MOLT-4 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 7 shows the activity data of the cellular RPMI-8226 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 8 shows the activity data of the cell a2780 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 9 shows the activity data of the OVCAR-3 viability assay for selected compounds of the invention. Suppression is indicated as IC50And using the following legend: a ═ IC50Less than 1 uM; b ═ IC50Greater than 1uM but less than 10 uM; c ═ IC50Greater than 10 uM.
Table 10 shows comparative data for a panel of assays showing CDK7 selectivity in the CDK family for compound 210 of the present invention.
Table 11 summarizes compounds 1-279 according to structure and corresponding features.
Figure 1 shows the in vivo anti-tumor activity of CDK7 inhibitors in an OVCAR-3 xenograft model.
Examples
The invention will now be further described with reference to the following examples, which are intended to illustrate, but not limit the scope of the invention.
Example 1: enzymatic assay for CDK1, CDK2, CDK5 and CDK7
Enzyme binding assay protocol for CDK1, CDK2, CDK5, and CDK7
In FRET-based
Figure BDA0002830360940000411
The inhibitory activity of the corresponding compounds on CDK kinases was tested at Km value of ATP in the Ultra kinase assay (Perkin Elmer) using ULightTMLabelled peptide substrates and suitable europiumA labeled anti-phosphoprotein antibody. Test compounds were prepared using DMSO solutions, then using an automated liquid manipulator (POD)TM810, Labcyte) 4-fold serial dilutions of 8 doses were prepared and 80 nL/well of the diluted compound solution was added to a 384-well plate (Greiner, catalog No. 784075). Then 68nM of ULight-MBP peptide (Perkin Elmer, cat. No. TRF0109-M) and 5 ul/well ATP (Sigma, cat. No. A7699) were added to the plates. After centrifugation at 1000rpm for 1min, the purified CDK/cyclin complexes were added using the following concentrations, respectively. To each respective plate were added 24uM CDK 1/cyclin B (Invitrogen, catalog No. PR4768C), 22uM CDK 2/cyclin a (Invitrogen, catalog No. PV6290), 10uM CDK5/p25(Invitrogen, catalog No. PR8543B) and 400uM CDK 7/cyclin H/MNAT1(Invitrogen, catalog No. PR6749B) for CDK1, CDK2, CDK5 and CDK 7. Incubation was performed at 23 ℃ for 60min, and then a mixture of Eu-labeled anti-phosphorylated myelin basic protein antibody (PE, Cat. TRF0201-M) and EDTA (Invitrogen, Cat. 15575038) in Lance detection buffer (Perkin Elmer, Cat. CR97100) was added to each well. After an additional incubation at 23 ℃ for 60min, the fluorescence of the test substances was measured using an Envision leader (Perkin Elmer, USA) [ laser as excitation light; APC615nm and europium 665 as first and second emission filters]. Data were analyzed using XL Fit software.
Example 2: cells HCT116, H460, MV4-11, MM.1S, MOLT-4, RPMI-8226, A2780 and OVCAR-3 Viability assay
Cell culture
Human T-cell acute lymphoblastic leukemia cell line MOLT-4(ATCC, catalog number CRL-1582), human multiple myeloma cell line RPMI-8226(Invitrogen, catalog number 22400-089) and MM.1S (ATCC, catalog number CRL-2974), NSCLC (non-small cell lung cancer) cell line H460(ATCC, catalog number HTB-177), human colorectal cancer cell line HCT116(ATCC, catalog number CCL-247), human acute monocytic leukemia cell line MV4-11(ATCC, catalog number CRL-9591), OVCAR-3(ATCC, catalog number HTB-161) and A2780(ECACC, catalog number 93112519) were obtained from ATCC. Cells were grown supplemented with 10% FBS (Invitrogen, Cat. No.)10099141) and 1% penicillin/streptomycin (Invitrogen, Cat. No. 15070063) in RPMI-1640 medium (Invitrogen, Cat. No. 22400-2And (5) culturing. All cell lines were routinely tested for mycoplasma.
Cellular HCT116, H460, MV4-11, MM.1S, MOLT-4, RPMI-8226, A2780 and OVCAR-3 viability assay protocol
To test the effect of CDK7 inhibitors on inhibiting the growth of target cancer cells, a 72 hour viability assay was performed. Briefly, candidate cell lines were plated in 96-well plates at the following cell densities, respectively. 1X10 for MOLT-4, RPMI-8226, MV4-11 and MM.1S4Individual cells/well, 5X 10 for H460, HCT116 and OVCAR-33And 1X10 for A27803. After 24 hours, cells were treated with various concentrations of compounds (ranging from 0.0015uM to 10 uM). DMSO solvent without compound served as control, and the final DMSO concentration was below 0.1%. At 37 deg.C, 5% CO2After 72 hours incubation in the incubator, the viability of the cells was analyzed using the CellTiter-Glo luminescent cell viability assay (Promega, Cat. No. G7570). All viability assays were performed in duplicate and luminescence was read using Envision (Perkin Elmer, USA). Data were analyzed using XLfit software.
50Example 3: in vitro IC profiles of CDK family kinases
In vitro IC of CDK7 inhibitors in 28 CDK family kinases50Situation(s)
The IC50 status of the compounds was determined by ProQinase GmbH (Freiburg, Germany) using 28 CDK family protein kinases. All procedures and materials are provided by ProQinase GmbH. Briefly, in the process, 90 μ l H was added to each well of a compound dilution plate2And O. To minimize potential precipitation, H was only introduced a few minutes before transferring the compound solution to the assay plate2O is added to the plate. The plates were shaken well to give "compound diluted plates/10% DMSO". The compound dilution plates were discarded at the end of the experimental day. For the assay (seeBelow), 5 μ Ι of solution from each well of the compound dilution plate was transferred to the assay plate. The final volume of the assay was 50. mu.l. All compounds were formulated at 1X10-05M to 3x 10-10The 10 final assay concentrations in the M range were tested. The final DMSO concentration in the reaction mixture was 1% in all cases. All protein kinases supplied by ProQinase were expressed in Sf9 insect cells or e.coli as recombinant GST fusion proteins or His-tagged proteins as full-length or enzymatically active fragments. All kinases were generated from human cDNA and purified by GSH affinity chromatography or immobilized metal affinity chromatography. The purity of the protein kinase was checked by SDS-PAGE/Coomassie staining and the identity by mass spectrometry. Kinases from the external suppliers Carna Biosciences inc, Invitrogen corp, and Millipore corp were expressed, purified, and quality controlled using the supplier's readings. Protein kinase assay using radiometry: (
Figure BDA0002830360940000431
Activity assay) to measure the kinase activity of the 28 protein kinase. All kinase assays were performed in 96-well FlashPlates from PerkinElmer (Boston, MA, USA)TMIn a 50. mu.l reaction volume. The reaction mixture was incubated at 30 ℃ for 60 minutes. With 50. mu.l of 2% (v/v) H3PO4The reaction was stopped, the plate was emptied and washed twice with 200. mu.l 0.9% (w/v) NaCl.33The incorporation of Pi was determined using a microplate scintillation counter (Microbeta, Wallac). All assays used BeckmanCoulter/SAGAINTMAnd (4) the core system. As part of the data evaluation, the low control value from a particular plate was subtracted from the high control value of the corresponding plate and all 80 "compound values". The residual activity (in%) of each well of a particular plate was calculated by the following formula: residual activity (%) ═ 100X [ (cpm of compound-low control)/(high control-low control)]. The results are presented in table (table 10).
Example 4: in vivo efficacy studies for OVCAR-3 model
In vivo efficacy of CDK7 inhibitors in OVCAR-3 human epithelial ovarian cancer xenograft model
To evaluate the inhibitory activity on the growth of OVCAR-3(ATCC, HTB-161) xenograft tumors, an in vivo efficacy study was conducted. OVCAR-3 cells were cultured in RPMI-1640 medium (Gibco, C2400500BT) supplemented with 20% fetal bovine serum (HyClone, SV30087.03), 0.01mg/ml bovine insulin (Yuanyue, S12033), and 1% antibiotic (Gibco, 15240-2Is grown in an air atmosphere. To establish tumors, 10X 10 was used6Individual OVCAR-3 cells were injected subcutaneously into the right upper flank of female Balb/c nude mice (Vital River Laboratory Animal co., ltd., Beijing) in 200 μ l PBS (Corning, 21-031-CVR) mixed with 50% matrigel (Corning, 354234). Tumor volumes were measured twice weekly and body weights were monitored daily. Tumor size of mice was measured in two dimensions using calipers and using the formula "V ═ 0.5a xb2"calculate tumor volume (mm)3) Where a and b are the long and short diameters of the tumor in mm, respectively. The animals were randomized into three groups of 8 animals each based on tumor volume. To assess efficacy, compound-210 was administered orally using 70% PEG400(Sigma-Aldrich, P3265) in distilled water as the vehicle. Up to about 160mm in mean tumor size3Thereafter, the animals were treated daily with vehicle, 20mg/kg or 40mg/kg of Compound-210 (q24h/qd schedule) to 25 days after randomization. Statistical analysis of tumor volume differences between groups was performed on the data obtained for PG-D25. All data were analyzed using Graphpad Prism (Graphpad, Prism 6.00). p is a radical of<0.05 was considered statistically significant. The results are shown in the figure (fig. 1).
Example 5: derivatization of the pyrazolo-triazines and pyrazolo-pyrimidines common skeleton
The proposed compounds were derivatized according to the methods outlined below (schemes 1-56). The resulting derivatives were examined for enzyme-binding cellular activity (HCT116, H460, MV4-11, MM.1S, MOLT-4, RPMI-8226, A2780, and OVCAR-3), CDK7 selectivity in the CDK family, and in vivo potency studies (OVCAR-3) using the assays described above (examples 1, 23, and 4), and the results are summarized in tables 1-10 and FIG. 1. The synthesized compounds 1-279 are shown in table 11.
Scheme 1-a: general synthetic route
Figure BDA0002830360940000451
Processes for preparing the compounds of I-3 and I-4 are shown in scheme 1-a.
Route I: compound C7 can be prepared by reacting in POCl3The synthesis is carried out by treatment in the presence of a catalyst. Compound C7 can be further treated with group B in the presence of DIPEA to afford compound I-1. Compound I-1 can be further treated with m-CPBA to provide a compound of formula I-2. Compounds of formula I-3 can be synthesized by treating compound I-2 with group A.
Route II: scheme II has similar reaction conditions and a different sequence than scheme I.
Compound C13 can be synthesized by treatment in the presence of m-CPBA. Compounds of formula II-1 can be synthesized by treating compound C13 with the appropriate group a. The compound II-2 can be prepared by reacting in POCl3The synthesis is carried out by treatment in the presence of a catalyst. Compounds of formula I-3 can be synthesized by treating compound II-2 with DIPEA and group B.
Deprotection step: compounds of formula I-4 can be prepared by using compounds of formula I-3 in the presence of an acid such as TFA, HBr and AcOH or a base such as hydrazine.
Scheme 1-b: general synthetic route
Figure BDA0002830360940000461
The methods for preparing the compounds of III-2 and III-3 are shown in scheme 1-b.
Route III: compound E1 can be synthesized by treatment in the presence of NaOEt. Compound E2 can be prepared by reacting in POCl3The synthesis is carried out by treatment in the presence of a catalyst. Compound E2 can be placed at K2CO3Exist ofFurther treatment with group B gave compound III-1. Compound III-1 can be further treated with group A to provide a compound of formula III-2. The compound of formula III-3 can be prepared by using the compound of formula III-2 in the presence of an acid such as TFA, HBr and AcOH or a base such as hydrazine.
Synthesis procedure for E1
Na (859mg, 37.4mmol) was added to anhydrous EtOH (100mL) at 10 deg.C, and the resulting mixture was stirred at 10 deg.C and N2After stirring under atmosphere for 1 hour, compound C3(3.90g, 31.1mmol) and diethyl malonate (5.99g, 37.4mmol) were added to the mixture. Mixing the mixture in N2Heating and stirring was continued at 80 ℃ under an atmosphere for 15 hours to give a yellow solution. TLC showed the reaction was complete. The reaction mixture was cooled to room temperature and concentrated to obtain a residue. The residue was dissolved in water (60mL) and 3M HCl was acidified to pH 3 and filtered to give compound E1(3.60g) as an off-white solid.
Synthesis procedure for E2
To compound E1(3.60g, 18.6mmol) in POCl3To a solution of (57.1g, 373mmol) was added N, N-diethylaniline (2.78g, 18.6 mmol). The resulting mixture is mixed with N2Heating and stirring was carried out at 100 ℃ for 2 hours under an atmosphere to obtain a red solution. TLC showed the reaction was complete and one major spot was formed. The reaction mixture was concentrated to remove most of the solvent and poured onto H2O (40mL) was extracted with DCM (50 mL. times.3). The organic layer was washed with brine (50 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product. The crude product was purified by column on silica gel to obtain compound E2(2.85g) as a yellowish solid.
General scheme for group A
Scheme 2: synthetic route to A1
Figure BDA0002830360940000471
Synthesis procedure of A1
A mixture of 4-aminophenol (1.00g, 9.16mmol) and 1, 4-dibromobutane (9.89g, 45.8mmol) in DMF (100mL) was stirred at 65 ℃ for 18 h. Saturated NaHCO3(150mL) was carefully added dropwise to the reaction mixture to quench the reaction. The mixture was diluted with EtOAc (100 mL). The organic phase was separated and the aqueous phase was extracted with EtOAc (100mL x2), the combined organic phases were washed with water (70mL x 4), brine (100mL) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product. The crude compound was purified by Combi-Flash to give compound a1(110mg) as a brown powder.
Scheme 3: synthetic route to A5
Figure BDA0002830360940000472
Synthesis procedure of A4
To compound A2(1.00g, 5.76mmol), compound A3(2.16g, 11.5mmol) and PPh3(3.02g, 11.5mmol) in dry THF (15mL) DEAD (2.01g, 11.5mmol) was added and the mixture was stirred at 20 ℃ for 17 h. TLC showed the reaction was complete. The reaction was quenched with water (50mL), extracted with EtOAc (50mL x3), and the combined extracts were washed with brine (100mL) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give 1.60g of compound a4 as a white powder.
Synthesis procedure of A5
To compound A4(800mg, 2.33mmol) and NH4A suspension of Cl (1.25g, 23.3mmol) in EtOH (15mL) was added Zn (1.53g, 23.3mmol) and the mixture was stirred at 50 ℃ for 17 h. TLC and LCMS showed the reaction was complete. The mixture was filtered, the filter cake was washed with DCM (50mL x2), and the combined organic phases were reduced pressureConcentration gave a residue, which was dissolved in DCM (100mL), washed with water (50mL × 3) and dried over anhydrous Na2SO4Above, dried, filtered, and the filtrate was concentrated under reduced pressure to obtain 650mg of compound a5 as a yellow gum.
Scheme 4: synthetic route to A9
Figure BDA0002830360940000481
Synthesis procedure of A8
To compound A6(1.00g, 5.78mmol) in H2To a solution of O (5mL) and dioxane (10mL) was added Compound A7(2.14g, 6.94mmol), K2CO3(2.00g, 14.5mmol) and Pd (dppf) Cl2(422mg, 0.578 mmol). Reacting the reaction mixture in N2Stirred under an atmosphere at 90 ℃ for 16 hours. LCMS showed 50.4% desired MS. The mixture was washed with DCM (50mL) and H2Partition between O (50 mL). The aqueous phase was extracted with DCM (50 mL). The combined organic extracts were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound A8(1.30g) as a white powder.
Synthesis procedure of A9
To a suspension of compound A8(1.30g, 4.72mmol) in MeOH (2mL) was added Pd/C (0.13g, 50% moisture content, 10% Pd) and the mixture was taken up in H2Stirring was carried out under a balloon (15psi) at 25 ℃ for 2 hours to give a black suspension. LCMS showed compound A8 was consumed and the desired MS was observed. The mixture was filtered and concentrated under reduced pressure to give compound a9(1.20g) as a yellow powder.
Scheme 5: synthetic route to A13
Figure BDA0002830360940000491
Synthesis procedure of A11
To Et3N (21mL) in MeNO2(50mL) Compound A10(5g, 25.1mmol) was added. The mixture was stirred at 10-15 ℃ for 48 hours to give a yellow suspension. The suspension was diluted with EtOAc (100mL) and saturated NH with water (100mL)4Cl (100mL), brine (100mL) and washed over Na2SO4Drying above, filtration and concentration under reduced pressure gave compound a11(7.6g) as a yellow solid.
Synthesis procedure of A12
To a mixture of compound A11(2g, 7.68mmol), imidazole (2.62g, 38.4mmol) in DMF (5mL) was added chloro (triethyl) silane (10 mL). The mixture was stirred at 70-80 ℃ for 12 hours to give a yellow mixture. The mixture was cooled to room temperature and diluted with water (50 mL). The aqueous phase was extracted with EtOAc (50mL x 3). The combined extracts were washed with brine (50mL) over Na2SO4Drying above, filtration and concentration under reduced pressure gave compound a12(5g) as a yellow oil.
Synthesis procedure of A13
To a mixture of Pd/C (1g, 10%) in MeOH (100mL) was added compound A12(4.9g, 13.08 mmol). The suspension is degassed under vacuum and treated with H2And purging for several times. Subjecting the mixture to conditions of 25 ℃ and H2Stirring (50psi) for 30 hours gave a black mixture. Crude HNMR showed the reaction was complete. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a yellow gum which was purified by Combi flash chromatography to give compound a13(2.3g) as a yellow oil.
Scheme 6: synthetic route to A16
Figure BDA0002830360940000492
Synthesis procedure of A15
A mixture of A14(1.5g, 9.54mmol), N-benzylhydroxylamine (2.28g, 14.3mmol), (HCHO) N (2.15g, 23.8mmol) and TEA (1.45g, 14.3mmol, 2.0mL) in toluene (100mL) was refluxed for 20 hours. Many white solids were observed. LCMS showed reaction completion. Most of the solvent was removed under reduced pressure. The residue was in EtOAc (50mL) and H2Partition between O (50 mL). The aqueous phase was extracted with EtOAc (50mL × 2). The organic extracts were washed with brine (100 mL. times.3) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give the crude product, which is purified by Combi flash chromatography to give compound a15(2.05g) as a pale yellow solid.
Synthesis procedure of A16
To a solution of A15(1.5g, 5.13mmol) in EtOH (10mL) was added Pd (OH)2C (0.5g, 20% purity). The reaction suspension is washed with H2(balloon, 15psi) purge several times and toluene at 15 ℃ for 16 hours to give a black mixture. TLC (PE/EA ═ 1/1) showed a new spot. The reaction was diluted with MeOH (200mL) and filtered over a pad of celite. The filtrate was concentrated under reduced pressure to give A16(1.15g, crude) as a white solid.
Scheme 7: scheme A18
Figure BDA0002830360940000501
Synthesis procedure of A18
To a mixture of A17(500mg, 5.61mmol) in DCM (10mL) was added Boc2O (1.35g, 6.17mmol), and the mixture was stirred at 25 ℃ for 16 hours to give a colorless oil. TLC showed the reaction was complete. The mixture was concentrated under reduced pressure to give compound a18(1.2g, crude) as a colorless oil.
Scheme 8: synthetic route to A21
Figure BDA0002830360940000502
Synthesis procedure of A20
To a solution of Compound A19(500mg, 2.46mmol) in DMF (5mL) was added NaN3(319mg, 4.92mmol) and NH4Cl (158mg, 2.95 mmol). The mixture was stirred at 80 ℃ for 3 hours to give a yellow mixture. LCMS showed reaction completion. The mixture was dissolved in EtOAc (30mL) and H2Partition between O (20 mL). The aqueous phase was extracted with EtOAc (30mL × 2). The combined organic extracts were washed with anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound a20(820mg, crude) as a yellow oil.
Synthesis procedure of A21
To a mixture of compound A20(820mg, 3.33mmol) in MeOH (10mL) was added Pd/C (100 mg). The mixture was stirred at 25 ℃ for 16 hours to give a black mixture. TLC and LCMS showed the reaction was complete. The mixture was filtered. The filtrate was concentrated under reduced pressure to give compound a21(680mg, crude) as a yellow oil.
Scheme 9: synthetic route to A26
Figure BDA0002830360940000511
Synthesis procedure of A24
To a mixture of A23(3.34g, 18.0mmol) and TMBAC (335mg, 1.80mmol) in IPA (50mL) was added A22(5.00g, 54.1mmol) and the mixture was stirred at 20 deg.C for 72 h to form a white mixture. TLC (eluent: PE/EtOAc ═ 2/1) showed a new spot. The mixture was dissolved in EtOAc (80mL) and H2Partition between O (80 mL). The aqueous phase was extracted with EtOAc (80mL × 2). The combined organic extracts were washed with brine (80 mL. times.3) over anhydrous Na2SO4Dried, filtered and concentrated under reduced pressure to give a residue which is purified by Combi flash chromatography (Combi flash) to give as a white solidBulk A24(1.1 g).
Synthesis procedure of A25
To a mixture of compound A24(1g, 4.92mmol) in THF (10mL) was added TEA (597mg, 5.91mmol, 822uL) and 2-hydroxy-2-methyl-propionitrile (502mg, 5.91mmol), and the mixture was stirred at 75 ℃ for 12 hours to give a yellow mixture. LCMS showed the reaction remaining. The mixture was stirred for a further 16 hours to give a brown mixture. TLC showed the reaction was complete. The mixture was dissolved in EtOAc (30mL) and H2Partition between O (20 mL). The aqueous phase was extracted with EtOAc (30mL × 2). The combined organic extracts were washed with anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product. The crude product was purified by Combi flash chromatography to give compound a25(450mg) as an off-white solid.
Synthesis procedure of A26
To a mixture of Compound A25(40mg, 0.17mmol) in EtOH (10mL) was added PtO2(4.0mg, 0.017mmol, 0.1eq) and HCl (0.05mL), and the mixture was stirred at 25 ℃ for 1 hour to give a black mixture. TLC showed the reaction was complete. The mixture was filtered and concentrated under reduced pressure to give compound a26(40mg, crude) as an off-white gum.
Scheme 10: synthetic route to A29
Figure BDA0002830360940000521
Synthesis procedure of A29
To a solution of compound A27(200mg, 0.979mmol), TEA (409uL) in DCM (5mL) was added CbzOSu (217mg, 0.870mmol) at 0-10 deg.C and the mixture was stirred at 20 deg.C for 1 hour to give a yellow mixture. TLC showed the reaction was complete. The mixture was quenched with water (100mL) and extracted with EtOAc (100mL x 3). The combined organic extracts were washed with water (100mL) and washed with Na2SO4Drying, filtering, and concentrating under reduced pressure to obtain yellowThe oil was washed with PE (10mL) to give compound a29(310mg) as a yellow oil.
Synthesis procedure of A30
Compound a29(290mg) was followed by the same procedure as B14 to obtain 270mg of compound a30 as a yellow gum.
Commercially available reagents are used in group A, for example tert-butyl piperidin-3-ylcarbamate, tert-butyl 4-aminopiperidine-1-carboxylate, piperidine-4-carboxamide, tert-butyl (4-methylpiperidin-4-yl) carbamate, tert-butyl piperidin-4-ylcarbamate, benzyl piperidin-4-ylcarbamate, piperidin-4-ol, piperazine, piperazin-2-one, tetrahydro-2H-pyran-4-amine, tert-butyl 3-methylpiperazine-1-carboxylate, N- (piperidin-4-yl) acetamide, 1-methylpiperidin-4-ol, ((4-hydroxypiperidin-4-yl) methyl) carbamate, tert-butyl (4-hydroxypiperidin-4-yl) carbamate, Tert-butyl (morpholin-2-ylmethyl) carbamate, tert-butyl 4- (aminomethyl) piperidine-1-carboxylate, (R) -tert-butyl 3-hydroxypiperidine-1-carboxylate, tert-butyl 4- (piperidin-4-yl) piperazine-1-carboxylate, (piperidin-4-ylmethyl) carbamate, 1,2,3, 4-tetrahydro-2, 6-naphthyridine, (4-methylpiperidin-4-yl) carbamate, ((4-fluoropiperidin-4-yl) methyl) carbamate, (S) -2- (piperidin-2-yl) ethan-1-ol, (S) -piperidin-2-ylmethanol, tert-butyl (4-fluoropiperidin-4-yl) carbamate, tert-butyl (S) -2- (piperidin-2-yl) ethan-1-ol, and (S) -piperidin-2-ylmethanol, (1S,4S) -4-aminocyclohexan-1-ol, (1r,4r) -4-aminocyclohexan-1-ol, 4-methoxycyclohex-1-amine, (tetrahydro-2H-pyran-4-yl) methylamine, morpholine, cyclohexa-1, 4-diol, (S) -3- (hydroxymethyl) piperazine-1-carboxylic acid tert-butyl ester, 2- (aminomethyl) morpholine-4-carboxylic acid tert-butyl ester, 1H-indol-4-amine, 4- (trifluoromethoxy) piperidine, 4-ethoxypiperidine, 4-isopropoxypiperidine, 4-methoxycyclohexa-1-amine, 4-isopropoxycyclohexa-1-amine, tert-butyloxy-2- (aminomethyl) morpholine-4-carboxylic acid, tert-butyloxy-piperidine, 1H-indol-4-amine, 4- (trifluoromethoxy) piperidine, 4-ethoxypiperidine, 4, 1H-pyrrol-3-amine and 4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester.
General scheme for group B
Scheme 11: synthetic route of B3
Figure BDA0002830360940000531
Synthesis procedure of B2
To a solution of compound B1(5.00g, 41.3mmol) and phenol (5.80g, 61.9mmol) in DMA (50mL) was added 18-crown-6 (1.10g, 4.13mmol) and K2CO3(11.4g, 82.6mmol) and the reaction mixture was stirred at 110 ℃ for 16h to give a brown mixture. LCMS showed reaction completion. Addition of H to the reaction mixture2O (50mL), the reaction mixture was extracted with EtOAc (50mL x3), and the combined organic phases were extracted with H2O (40 mL. times.2) and brine (100mL) in anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a brown oil which is purified by Combi flash chromatography to give compound B2(9.80g) as a yellow oil.
Synthesis procedure of B3
To a solution of compound B2(1.00g, 5.12mmol) in MeOH (30mL) were added Raney-Ni (43.9mg, 0.512mmol) and NH3.H2O (3mL), the reaction mixture was in H2Balloon (15psi) and 15 ℃ for 16 hours to obtain a black suspension. TLC showed the reaction was complete. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to remove MeOH. The residue was diluted with DCM (20mL) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B3(820mg) as a yellow oil.
Scheme 12: synthetic route of B7
Figure BDA0002830360940000541
Synthesis procedure of B6
To a solution of 2-hydroxybenzonitrile B4(200mg, 1.68mmol) and 2-chloroquinoline B5(261mg, 1.60mmol) in DMA (3.0mL) was added Cs2CO3(1.04g, 3.20 mmol). The reaction mixture was stirred at 100 ℃ for 5 hours. TLC showed the reaction was complete. The residue was partitioned between water (20mL) and EtOAc (20 mL). The organic layer was washed with water (10 mL. times.2), brine (10mL),dried over anhydrous Na2SO4 and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound B6(80mg) as a yellow powder.
Synthesis procedure of B7
Compound B6(80mg) was followed by the same procedure as B3 to obtain 64mg of compound B7 as a yellow powder.
Scheme 13: synthetic route of B10
Figure BDA0002830360940000542
Synthesis procedure of B9
To a mixture of NaH (198mg, 4.96mmol, 60% in mineral oil) in THF (3mL) was added dropwise a solution of compound B8(571mg, 4.96mmol) in THF (3 mL). After the reaction mixture was stirred for 5 minutes, 2-fluorobenzonitrile (500mg, 4.13mmol) was added to the mixture. The reaction mixture was stirred at 40 ℃ for 2 hours. LCMS showed reaction completion. The reaction mixture was quenched with water (10mL) and extracted with DCM (10mL × 2). The organic layer was washed with water (10mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by Combi flash chromatography to give compound B9(432mg) as a light brown oil.
Synthesis procedure of B10
Compound B9(430mg) was followed the same procedure as B102 to obtain 410mg of compound B10 as a white powder.
Scheme 14: synthetic route of B14
Figure BDA0002830360940000551
Synthesis procedure of B13
To compound B11(150mg, 0.675mmol) and compound B12(114mg, 0.81 mmol)l) solution in DCM (10mL) DIPEA (174mg, 1.35mmol) was added. The resulting mixture was stirred at 20 ℃ for 12 hours to give a yellowish solution. TLC showed the reaction was complete and one major spot was formed. The reaction mixture is purified by addition of H2Quench O (30mL) and extract with DCM (30 mL. times.2). The combined organic layers were washed with brine (10mL) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to obtain compound B13(200mg) as a white solid.
Synthesis procedure of B14
To a solution of compound B13(200mg, 0.613mmol) in DCM (7mL) was added TFA (3 mL). The resulting mixture was stirred at 20 ℃ for 1 hour to give a yellow solution. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give compound B14(130mg) as a yellow oil.
Scheme 15: synthetic route of B18
Figure BDA0002830360940000561
Synthesis procedure of B17
To a solution of compound B15(1.00g, 3.49mmol) and compound B16(536mg, 4.19mmol) in DME (10mL) was added H2Pd (dppf) Cl in O (2.5mL)2(128mg, 0.174mmol) and Na2CO3(370mg, 3.49mmol) and the reaction mixture was stirred at 90 ℃ for 3 h to give a black suspension. TLC showed the reaction was complete. Adding H to the reaction solution2O (10mL), extracted with EtOAc (10mL x2), and the organic layer was washed with brine (10mL) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give a brown oil which was purified by Combi flash chromatography to give compound B17(705mg) as a yellow oil.
Synthesis procedure of B18
Compound B17(350mg) was followed by the same procedure as B14 to obtain 220mg of compound B18 as a yellow powder.
Scheme 16: synthetic route of B21
Figure BDA0002830360940000562
Synthesis procedure of B20
Compound B19(2.00g, 16.5mmol), pyrrolidine (1.29g, 18.1mmol), K2CO3A mixture of (4.56g, 33.0mmol) in DMF (10mL) was stirred at 60 ℃ for 16 h. TLC showed the reaction was complete. The mixture was poured into water (100 mL). The mixture was extracted with EtOAc (30mL x3), and the combined mixture was washed with water (50mL x2), brine (50mL x2) over anhydrous Na2SO4The above was dried, filtered and concentrated under reduced pressure to give compound B20(2.78g) as a colorless oil.
Synthesis procedure of B21
Compound B20(1.5g) was followed by the same procedure as B3 to obtain 1.5g of compound B21 as a colorless oil.
Scheme 17: synthetic route of B24
Figure BDA0002830360940000571
Synthesis procedure of B23
To compound B22(5g, 35.7mmol) and Cs2CO3(29.1g, 89.2mmol) in DMF (50mL) was added 2-fluorobenzonitrile (6.48g, 53.5mmol, 5.69 mL). The reaction mixture was stirred at 25 ℃ for 16 hours to give a yellow mixture. TLC showed the reaction was complete. The reaction mixture is purified by addition of H2O (200mL) was quenched and extracted with EtOAc (150mL x 2). The combined organic layers were washed with Na2SO4Dried, filtered and concentrated under reduced pressure to give a crude productA compound (I) is provided. The crude product was purified by silica gel column to give compound B23(5g) as a white powder.
Synthesis procedure of B24
At 0 ℃ and N2To a solution of compound B23(200mg, 0.829mmol) in THF (2mL) under atmosphere was added LiAlH4(126mg, 3.32 mmol). The reaction mixture was stirred at 25 ℃ for 2 hours to give a yellow solution. LCMS showed reaction completion. The reaction was quenched slowly with water (1mL) and aqueous NaOH (1mL, 2.0M) at 5 ℃. The mixture was stirred at 5 ℃ for 10 minutes. The mixture was filtered and the filtrate extracted with EtOAc (5mL × 2). The combined organic phases were washed with brine (5mL) over anhydrous Na2SO4Above was dried, filtered and concentrated to give crude compound B24(180mg) as a pale yellow gum.
Scheme 18: synthetic route of B28
Figure BDA0002830360940000581
Synthesis procedure of B26
At 0-5 ℃ and N2To a solution of compound B25(5g, 35.7mmol) in THF (50mL) under atmosphere was added MeMgBr (3M, 47.6mL) dropwise. The resulting mixture was stirred at 10 ℃ for 15 hours to form a white suspension. TLC showed the reaction was complete. The mixture was poured over saturated NH4Aqueous Cl solution (50 mL). The mixture was extracted with EtOAc (50mL x 3). The combined organic extracts were washed with brine (80 mL. times.2) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B26(2.6g) as a white solid.
Synthesis procedure of B27
To a mixture of compound B26(2.6g, 20.6mmol) and 2-fluorobenzonitrile (3.00g, 24.7mmol) in DMF (50mL) was added Cs2CO3(13.4g, 41.2 mmol). The mixture was stirred at 15 ℃ for 15 hours to form a white suspension. TLC displayIndicating that the reaction is complete. The mixture was dissolved in EtOAc (150mL) and H2Partition between O (150 mL). The aqueous phase was extracted with EtOAc (150mL × 2). The combined organic extracts were washed with brine (100 mL. times.3) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give the crude product, which is purified by Combi flash chromatography to give compound B27(3.6g) as a white solid.
Synthesis procedure of B28
In N2To a solution of compound B27(3.6g, 15.84mmol) in MeOH (100mL) under atmosphere was added Raney-Ni (1g in water). The suspension is degassed under vacuum and treated with H2And purging for several times. Mixing the mixture in H2(15psi) and stirred at 20 ℃ for 15 hours to form a black suspension. TLC showed the reaction was complete. The reaction mixture was filtered over a pad of celite. The filter cake was washed with MeOH (80 mL). The filtrate was concentrated under reduced pressure to give compound B28(3.5g) as a colorless gum.
Scheme 19: synthetic route of B31
Figure BDA0002830360940000591
Synthesis procedure of B30
To a mixture of morpholine (2.63g, 30.2mmol) and compound B29(500mg, 2.02mmol) in toluene (5mL) was added XPhos (192mg, 0.403mmol), Cs2CO3(1.64g, 5.04mmol) and Pd2(dba)3(185mg, 0.202mmol) and the reaction mixture was stirred under N2Stirring was carried out at 110 ℃ for 16 hours under an atmosphere to give a black suspension. LCMS (Rt ═ 0.908min) showed the reaction was complete. The reaction mixture was partitioned between EtOAc (80mL) and water (80 mL). The aqueous phase was extracted with EtOAc (70mL x 2). The combined organic layers were washed with water (100mL x2), brine (100mL x2) and washed over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The reaction mixture was purified by Combi flash chromatography to give the compound as a brown gumSubstance B30(190 mg).
Synthesis procedure of B31
Compound B30(190mg) was followed by the same procedure as B3 to obtain 170mg of compound B31 as a colorless gum.
Scheme 20: synthetic route of B38
Figure BDA0002830360940000592
Synthesis procedure of B33
To a mixture of compound B31(10g, 37.7mmol) in DMF (5mL) was added compound B32(9.13g, 75.3 mmol). The mixture was stirred at 50 ℃ for 12 hours to give a yellow suspension. LCMS showed reaction completion. Water (30mL x2) was added to the mixture, which was filtered and concentrated under reduced pressure to give a yellow powder, which was washed with PE (50mL) to give compound B32(7g) as a brown powder.
Synthesis procedure of B34
To a mixture of compound B33(4g, 18.7mmol) in MeOH (30mL) was added NH4Cl (9.99g, 186mmol) and Zn (12.2g, 186 mmol). The mixture was stirred at 25 ℃ for 15 hours to give an off-white suspension. LCMS showed reaction completion. After filtration, the filter cake was washed with MeOH (50mL) and the filtrate was partitioned between DCM (50mL) and water (50 mL). The aqueous phase was extracted with DCM (50mL x2), the combined extracts were washed with water (50mL x2) over anhydrous Na2SO4Dried above and concentrated under reduced pressure to give compound B34(2g) as a yellow powder.
Synthesis procedure of B36
To a mixture of compound B34(2g, 10.8mmol) in n-BuOH (20mL) was added compound B35(1.94g, 10.8 mmol). The reaction was refluxed at 117 ℃ for 48 hours to give a yellow suspension. LCMS showed reaction completion. To the mixture were added NaOH (868mg, 21.7mmol) and H2O (10mL), dioxane (10mL), tert-butoxycarbonyl tert-butyl carbonate (3.56g,16.2 mmol). The mixture was stirred at 25 ℃ for 2 hours to give a yellow suspension. LCMS (Rt ═ 1.268min) showed the reaction was complete. The mixture was partitioned between DCM (50mL) and water (50 mL). The aqueous phase was extracted with DCM (50mL x2), the combined extracts were washed with water (50mL x2) over anhydrous Na2SO4Dry above and concentrate under reduced pressure to give a yellow gum which is purified by Combi flash chromatography to give compound B36(400mg) as a yellow oil.
Synthesis procedure of B37
Compound B36(1.0g) was followed by the same procedure as B72 to obtain 400mg of compound B37 as a yellow solid.
Synthesis procedure of B38
Compound B37(200mg) was followed by the same procedure as B3 to obtain 200mg of compound B38 as a yellow powder.
Scheme 21: synthetic route of B42
Figure BDA0002830360940000611
Synthesis procedure of B41
To compound B39(423mg, 2.88mmol), compound B40(500mg, 2.40mmol), K2CO3(663mg, 4.80mmol) in dioxane (5mL)/H2Mixture in O (1mL) Pd (dppf) Cl was added2(175mg, 0.24 mmol). The mixture was stirred at 110 ℃ for 16 hours. TLC showed a new spot. The mixture was poured into water (20 mL). The mixture was extracted with DCM (30mL × 3). The combined mixture was washed with water (50mL x2) over anhydrous Na2SO4Dry above then filter and concentrate under reduced pressure to give a residue (brown gum). The residue was purified by Combi flash chromatography to give compound B41(200mg) as a red powder.
Synthesis procedure of B42
Compound B41(200mg) was followed by the same procedure as B3 to obtain 200mg of compound B42 as a brown oil.
Scheme 22: synthetic route of B46
Figure BDA0002830360940000612
Synthesis procedure of B45
A solution of compound B43(300mg, 1.37mmol) and compound B44(335mg, 1.51mmol) in i-PrOH (10mL) was heated at 80 ℃ and stirred for 4h, then tributylphosphine (1.39g, 6.85mmol) was added to the mixture and stirring continued for 12h to give a yellow solution. LCMS and TLC showed the reaction was complete. The reaction mixture is purified by addition of H2O (50mL) was quenched and extracted with EtOAc (50 mL. times.2). The combined organic layers were washed with brine (20 mL. times.2) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound B45(524mg) as a yellow solid.
Synthesis procedure of B46
Compound B45(524mg) was followed B14 and the same procedure to obtain 430mg of compound B46 as a yellow solid.
Scheme 23: synthetic route of B50
Figure BDA0002830360940000621
Synthesis procedure of B49
To a solution of Compound B47(500mg, 3.70mmol) in AcOH (10mL) was added Compound B48(1.11g, 4.44mmol), Pd (OAc)2(415mg, 1.85mmol), and then subjecting the reaction mixture to O at 25 deg.C2Stirring (15psi) for 16 hours gave a black suspension. LCMS and TLC showed the reaction was complete. The reaction was poured into water (50mL) and in EtOAc (100mL) and water (100 m)L) are distributed. The aqueous phase was extracted with EtOAc (100mL x 2). The combined organic layers were washed with water (100 mL. times.2), brine (100mL) and washed with anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound B49(240mg, 0.705mmol) as a yellow gum.
Synthesis procedure of B50
Compound B49(240mg) was followed by the same procedure as B14 to obtain 170mg of compound B50 as a yellow gum.
Scheme 24: synthetic route of B56
Figure BDA0002830360940000622
Synthesis procedure of B53
To a solution of compound B51(600mg, 3.35mmol) in i-PrOH (10mL) was added compound B52(744mg, 3.35 mmol). The reaction solution was heated to 80 ℃ and stirred for 3 hours to give a yellow solution. The reaction was cooled to 30-40 ℃, tributylphosphine (2.03g, 2.48mL) was added and stirring was continued at 80 ℃ for 16 hours to give a black solution. TLC showed the reaction was complete. The reaction mixture is purified by addition of H2O (100mL) was quenched and extracted with EtOAc (100 mL. times.2). The combined organic layers were washed with saturated NH4Cl (50mL) and brine (10 mL. times.2) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound B53(784mg) as a yellow solid.
Synthesis procedure of B54
To compound B53(784mg, 2.06mmol) in MeOH (5mL) and H2Solution in O (1mL) LiOH. H was added2O (431mg, 10.3 mmol). The resulting mixture was stirred at 20 ℃ for 12hrs to give a yellow suspension. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the reaction mixtureMeOH, then acidified to pH-4 with 1M HCl, precipitated a large amount of white solid, and the mixture was filtered under reduced pressure to give compound B54(724mg) as a white solid, a filter cake.
Synthesis procedure of B55
To a solution of compound B54(625mg, 1.70mmol) and methylamine (230mg, 3.40mmol) in DMF (10mL) was added Et3N (516mg,5.10mmol) and HATU (647mg, 1.70mmol, 1 eq). The resulting mixture was stirred at 20 ℃ for 2hr to give a yellow solution. TLC showed the reaction was complete and a major new spot was formed. Subjecting the reaction mixture to hydrogenation with H2O (20mL) quenched and precipitated a large amount of white solid, which was then filtered under reduced pressure to give Compound B55(693mg) as a white solid.
Synthesis procedure of B56
Compound B55(200mg) was followed by the same procedure as B14 to obtain 130mg of compound B56 as a yellow oil.
Scheme 25: 61 synthetic route
Figure BDA0002830360940000641
Synthesis procedure of B59
Compound B57(500mg) was followed by the same procedure as B41 to obtain 840mg of compound B59 as a yellow solid.
Synthesis procedure of B60
In N2To a solution of compound B59(350mg, 1.08mmol) in MeOH (10mL) under atmosphere was added Pd/C (20mg, 10% purity, wet weight). The suspension is degassed under vacuum and treated with H2And purging for several times. Reacting said mixture in H2(50psi) and stirred at 20 ℃ for 12h to give a black suspension. LCMS showed no reaction and acetic acid (0.1mL) was added to the reaction. The suspension is degassed under vacuum and treated with H2And purging for several times. Reacting said mixture in H2(50psi) and stirring at 20 ℃ for 12 hoursTo obtain black suspension. LCMS showed reaction completion. The reaction mixture was filtered through a pad of celite and concentrated under reduced pressure to give compound B60(450mg, crude) as a pale yellow oil, which was used in the next step without purification.
Synthesis procedure of B61
Compound B60(450mg) was followed by the same procedure as B14 to obtain 330mg of compound B61 as a yellow oil.
Scheme 26: synthetic route of B66
Figure BDA0002830360940000642
Synthesis procedure of B63
A solution of compound B62(1.00g, 8.84mmol) in DMF/DMA (20mL) was heated at 110 ℃ and stirred for 2h to give a red solution. The reaction mixture was concentrated under reduced pressure to give the crude product compound B63(1.20g) as a red solid.
Synthesis procedure of B64
To a solution of compound B63(700mg, 4.16mmol) in EtOH (10mL) was added NH2NH2.H2O (208mg, 4.16 mmol). The resulting mixture was heated at 100 ℃ and stirred for 16hrs to give a yellow solution. The reaction mixture was concentrated under reduced pressure to give compound B64(550mg) as a red solid.
Synthesis procedure of B65
Compound B64(400mg) was followed by the same procedure as B33 to obtain 445mg of compound B65 as a yellow solid.
Synthesis procedure of B66
Compound B65(100mg) was followed by the same procedure as B3 to obtain 91mg of compound B66 as a white powder.
Scheme 27: synthetic route of B70
Figure BDA0002830360940000651
Synthesis procedure of B69
To compound B67(300mg, 1.41mmol), compound B68(710mg, 2.83mmol), Cs2CO3(1.15g, 3.54mmol) in dioxane (3mL), H2Mixture in O (0.9mL) Pd (dppf) Cl was added2(104mg, 0.141 mmol). Subjecting the mixture to conditions of 100 ℃ and N2Stirring was carried out under an atmosphere for 12 hours to give a brown suspension. LCMS showed reaction completion. The mixture was cooled to room temperature and partitioned between DCM (50mL) and water (50 mL). The aqueous phase was extracted with DCM (30mL × 2). The combined extract phases were washed with water (30 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product as a red oil, which was purified by Combi flash chromatography to give compound B69(108mg) as a brown oil.
Synthesis procedure of B70
To a mixture of compound B69(100mg, 0.296mmol) in DCM (4mL) was added TFA (1 mL). The mixture was stirred at 15 ℃ for 20 minutes to give a yellow mixture. LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give compound B70(90mg) as a yellow oil.
Scheme 28: synthetic route of B75
Figure BDA0002830360940000661
Synthesis procedure of B72
To a mixture of compound B71(1g, 5.68mmol) in DCM (5mL) was added Boc2O (1.49g, 6.82 mmol). The mixture was stirred at 25 ℃ for 2 hours to give a yellow mixture. LCMS showed reaction completion. The mixture was partitioned between DCM (50mL) and water (50 mL). The aqueous phase was extracted with DCM (30 mL. times.2) and the mixture was combinedThe combined extracts were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B72(950mg) as a white powder.
Synthesis procedure of B74
To compound B72(850mg, 3.08mmol), compound B73(804mg, 3.08mmol) and Cs2CO3(2.51g, 7.69mmol) in dioxane (5mL) and H2Mixture in O (1.5mL) Pd (dppf) Cl was added2(225mg, 0.308mmol), and the mixture was heated at 100 ℃ and N2Stirring was carried out under an atmosphere for 12 hours to obtain a red mixture. LCMS showed reaction completion. The mixture was cooled to room temperature and partitioned between DCM (50mL) and water (50 mL). The aqueous phase was extracted with DCM (50mL × 2). The combined extract phases were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product, which is purified by Combi flash chromatography to give compound B74(420mg) as a white powder.
Synthesis procedure of B75
Compound B74(230mg) was followed by the same procedure as B14 to obtain 170mg of compound B75 as a yellow oil.
Scheme 29: synthetic route of B81
Figure BDA0002830360940000671
Synthesis procedure of B77
To a mixture of compound B76(8.00g, 56.3mmol) in MeOH (60mL) was added TEA (11.4g, 113mmol) and methylhydrazine (8.11g, 56.3 mmol). The mixture was stirred at 60 ℃ for 15 hours to form a brown mixture. The desired MS value was detected by LC-MS. The mixture was cooled to 5 ℃ and allowed to stand for 2 hours until white crystals precipitated, filtered, washed with ethanol and dried to give compound B77(3.5g) as a white solid.
Combination of B78Procedure for formation
A mixture of Compound B77(750mg, 4.80mmol) and TEA (1.34mL) in DCM (5mL) was cooled to 0 ℃ in a brine/ice bath. Then Tf was added dropwise2O (2.71g, 9.61mmol) while maintaining the temperature at 0 ℃. After the addition was complete, the reaction mixture was warmed to 20 ℃ and stirred for 1 hour to form a colorless mixture. LCMS showed complete consumption of the starting material. The reaction mixture was quenched with water (30mL) and the layers separated. The aqueous phase was extracted with DCM (50mL × 2). The combined organic extracts were washed with brine (80mL x3) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue which is purified by Combi flash chromatography to give compound B78(530mg) as a yellow oil.
Synthesis procedure of B80
To compound B78(430mg, 1.49mmol) and compound B79(544mg, 2.24mmol) in dioxane (4mL) and H2Mixture in O (1mL) with addition of Cs2CO3(1.22g, 3.73mmol) and Pd (dppf) Cl2(109mg, 0.15mmol), and the mixture was stirred under N2Stirring was carried out at 80 ℃ for 12 hours under an atmosphere to form a brown mixture. LCMS showed reaction completion. The mixture was filtered over a pad of celite. The filtrate was washed with EtOAc (30mL) and H2Partition between O (30 mL). The aqueous phase was extracted with EtOAc (30mL × 2). The combined organic extracts were washed with brine (80 mL. times.3) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give a residue which is purified by Combi flash chromatography to give compound B80(200mg) as a white solid.
Synthesis procedure of B81
To a mixture of methyl compound B80(150mg, 0.58mmol) in THF (4mL) at 0 deg.C was added LiAlH4(111mg, 2.94mmol) and the mixture was stirred at 10 ℃ for 12h to form a white mixture. LCMS showed reaction completion. The mixture is saturated with NH4Cl (10 mL). The mixture was filtered over a pad of celite and washed with EtOAc (10 mL). The filtrate was taken up in EtOAc (30 m)L) and H2Partition between O (30 mL). The aqueous phase was extracted with EtOAc (30mL × 2). The combined organic extracts were washed with brine (50 mL. times.3) over anhydrous Na2SO4Above was dried, filtered and concentrated under reduced pressure to give compound B81(100mg) as a white solid, which was used in the next step without purification.
Scheme 30: synthetic route of B88
Figure BDA0002830360940000681
Synthesis procedure of B84
To compound B82(1g, 7.14mmol), Cs2CO3(5.81g, 17.8mmol) in DMF (15mL) Compound B83(6.14g, 35.68mmol) was added. The mixture was stirred at 20 ℃ for 12 hours to give a yellow suspension. TLC showed the reaction was complete. The mixture was partitioned between EtOAc (100mL) and water (100mL), the aqueous phase was extracted with EtOAc (80mL x2), the combined extract phases were washed with water (80mL) and washed over Na2SO4Dry above, filter and concentrate under reduced pressure to give a yellow oil which is purified by Combi flash chromatography to give compound B84(1.9g) as a white solid.
Synthesis procedure of B85
To compound B84(300mg, 1.09mmol) in THF (2mL), MeOH (2mL), H2To the mixture in O (1mL) was added LiOH (130mg, 5.44 mmol). The mixture was stirred at 15 ℃ for 12 hours to give a yellow mixture. LCMS showed reaction completion. The mixture was concentrated under reduced pressure to remove MeOH. The aqueous phase was diluted with water (30mL), acidified to pH 4-5 with HCl (3M) and freeze dried to give compound B85(201mg) as a white solid.
Synthesis procedure of B87
To a mixture of compound B85(200mg, 0.808mmol), HOBt (131mg, 0.969mmol), EDCI (186mg, 0.969mmol) in DMF (3mL) was added DIEA (313mg, 2.42mmol), compound B86(32 mg, 2.42mmol)4mg, 1.62 mmol). The mixture was stirred at 20 ℃ for 12 hours to give a yellow mixture. LCMS showed reaction completion. Subjecting the reaction mixture to reaction with H2O (20mL) quenched and precipitated a large amount of white solid, which was then filtered under reduced pressure to give compound B87(205mg) as a white powder.
Synthesis procedure of B88
Compound B87(200mg) was followed by the same procedure as B3 to obtain 254mg of compound B88 as a yellow solid.
Scheme 31: synthetic route of B95
Figure BDA0002830360940000691
Synthesis procedure of B90
To a solution of compound B89(5g, 41.3mmol), pyridine (8.33mL) in DCM (50mL) was added TFAA (7.17mL) at 0 ℃. The mixture was stirred at 20 ℃ for 12 hours to give a yellow mixture. LCMS showed reaction completion. The reaction mixture was poured into 0.5N HCl (30mL) and stirred vigorously for 5 min. Separating the layers, and using CH for the aqueous layer2Cl2(3X 10 mL). The combined organic layers were washed with 0.5N HCl (20mL), H2O (2X 20mL) and saturated NaHCO3(20mL) washed and washed over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B90(9.3g, crude) as a yellow gum.
Synthesis procedure of B91
At 0 ℃ to CF3SO3H (2.44mL) solution in DCM (50mL) was added HNO3(622uL) and stirred at 0 ℃ for 30 minutes. The mixture was cooled to-70 ℃ and a solution of compound B90(3.00g, 13.8mmol) in DCM (20mL) was added for 1 h. The mixture was then stirred at-70 ℃ for 30 minutes. The mixture was stirred at-40 ℃ for 12 hours to give a yellow mixture. LCMS showed reaction completion. The orange-yellow reaction mixture was poured into ice (50g) and stirred vigorously for 10 minutes. To carry outSeparating the layers, and using CH for the aqueous layer2Cl2(3X 25 mL). Combining the organic layers with H2O (3X 50mL), saturated NaHCO3(50mL) and H2O (50mL) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B91(3.12g) as a yellow solid.
Synthesis procedure of B92
To a mixture of compound B91(500mg, 1.91mmol) in EtOH (10mL) was added Pd/C (0.1g, 10% purity, 50% water). The suspension is degassed under vacuum and treated with H2And purging for several times. Subjecting the mixture to conditions of 15 ℃ and H2Stirring was carried out under an atmosphere (15psi) for 12 hours to give a black suspension. LCMS showed reaction completion. The combined batch mixture was filtered and the filtrate was concentrated under reduced pressure to give compound B92(480mg) as a yellow oil.
Synthesis procedure of B94
To a solution of compound B92(390mg, 2.58mmol) in i-PrOH (15mL) was added compound B93(600mg, 2.58 mmol). The reaction solution was heated to 80 ℃ and stirred for 3 hours to give a yellow solution. The reaction was cooled to 30-40 ℃ and P (n-Bu) was added3(1.57g, 7.74mmol) and stirring at 80 ℃ was continued for 12h to give a dark brown solution. LCMS showed reaction completion. The reaction mixture is purified by addition of H2O (100mL) was quenched and extracted with EtOAc (100 mL. times.2). The combined organic layers were washed with saturated NH4Cl (50mL) and brine (10 mL. times.2) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue which is purified by Combi flash chromatography to give compound B94(200mg) as a yellow gum.
Synthesis procedure of B95
To a solution of compound B94(200mg, 0.600mmol) in MeOH (8mL) was added K2CO3(415mg, 3.00mmol) and the resulting mixture was stirred at 60-70 ℃ for 2h to give a yellow solution. TLC showed the starting material was consumed. The reaction mixture was concentrated under reduced pressure to giveCompound B95(141mg) as a yellow gum.
Scheme 32: synthetic route of B98
Figure BDA0002830360940000711
Synthesis procedure of B97
To a solution of compound B96(4.00g, 32.8mmol) in DMA (50mL) was added 1H-pyrazole (2.68g, 39.3mmol) and Cs2CO3(10.7g, 32.8 mmol). The reaction mixture was stirred at 80 ℃ for 5 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between water (250mL) and EtOAc (250 mL). The organic layer was washed with water (100 mL. times.2), brine (10mL) and washed with anhydrous Na2SO4Dried above and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound B97(4.76g) as a white powder.
Synthesis procedure of B98
To a solution of compound B97(1.3g, 7.64mmol) in MeOH (30mL) was added Raney-Ni (497mg, 5.81 mmol). Reacting the reaction mixture in H2Stirring was carried out under an atmosphere (15psi) at 20 ℃ for 3 hours. TLC showed the reaction was complete. The reaction mixture was filtered through a pad of celite. The filtrate was concentrated under reduced pressure to give 560mg of a dark brown gum as a crude product. The crude product was purified by Combi flash chromatography to give compound B98(350mg) as a light purple oil.
Scheme 33: synthetic route of B102
Figure BDA0002830360940000721
Synthesis procedure for B100
To compound B99(1.00g, 8.05mmol) and CuBr at 0 deg.C2(2.16g, 9.66mmol) in CH3Addition of Tert-butyl to CN (10mL)Butyronitrile (1.25g, 12.1mmol), and the reaction mixture was stirred at 15 ℃ for 3 hours to give a brown solution. TLC showed the reaction was complete. To the reaction mixture was added 2M HCl (50mL), extracted with EtOAc (30mL x3), and the organic phase was washed with brine (50mL) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a brown oil. The product was purified by Combi flash chromatography to give compound B100(380mg) as a yellow oil.
Synthesis procedure for B101
To a solution of compound B100(380mg, 2.02mmol) in dioxane (6mL) was added CuI (115mg, 0.606mmol), K2CO3(559mg, 4.04mmol), trans-N1,N2-dimethylcyclohexane-1, 2-diamine (86.2mg, 0.606mmol) and 1H-pyrazole (165mg, 2.42mmol), the reaction was stirred at 80 ℃ for 16H to give a brown suspension. TLC showed a new spot. 1H-pyrazole (500mg) was added to the reaction solution, and the solution was stirred at 110 ℃ for 8 hours. TLC showed the reaction was not complete. The mixture was filtered. The filtrate was partitioned between EtOAc (20mL) and water (20 mL). 28% NH for organic layer3.H2O (30mL x2), brine (20mL x2), over anhydrous Na2SO4Dry above then filter and concentrate under reduced pressure to give a yellow oil. The product was purified by Combi flash chromatography to give compound B101(180mg) as a white solid.
Synthesis procedure of B102
Compound B102(180mg) was followed by the same procedure as B3 to obtain 143mg of compound B101 as a yellow oil.
Scheme 34: synthetic route to B105
Figure BDA0002830360940000731
Synthesis procedure of B104
To compound B103(1.00g, 4.81mmol) and Pd(PPh3)4(556mg, 0.481mmol) mixture in anhydrous DMF (10mL) Zn (CN) was added2(678mg, 5.77 mmol). The reaction mixture is stirred under N2Heating was carried out at 80 ℃ for 16 hours under an atmosphere. LCMS showed reaction completion. The reaction mixture was cooled to room temperature, then the mixture was poured into water (50mL) and the crude product was extracted with EtOAc (100mL), the organic layer was washed with water (50mL x2), brine (50mL), over anhydrous Na2SO4Dry above, filter, and concentrate under reduced pressure to afford the crude product as a brown oil. The crude product was purified by Combi flash chromatography to give compound B104(700mg) as a white powder.
Synthesis procedure for B105
Compound B104(700mg) was followed by the same procedure as B102 to obtain 660mg of compound B105 as a brown oil.
Scheme 35: synthetic route to B111
Figure BDA0002830360940000732
Synthesis procedure for B107
To a mixture of Compound B106(10.0g, 45.4mmol) in DMF (5mL) was added MeNH2(2M in THF, 68.2mL), then K was added2CO3(9.42g, 68.2mmol) and the resulting mixture was stirred at 25 ℃ for 12h to give a yellow suspension. TLC showed the reaction was complete. The mixture was diluted with EtOAc (300mL), washed with water (200 mL. times.3) and brine (200mL) over anhydrous Na2SO4The above was dried, filtered and concentrated under reduced pressure to give compound B107(13.0g) as a yellow oil, which was used in the next step without purification.
Synthesis procedure for B108
To a mixture of compound B107(6.00g) in MeOH (60mL) was added AcOH (15.6g, 259mmol) followed by Fe powder (7.25g, 129.9mmol) and the resulting mixture was stirred at 25Stirring at deg.C for 12 hr. Crude LCMS showed the reaction progressed well. The suspension was filtered and washed with MeOH (80 mL). The filtrate was concentrated under reduced pressure. The residue was taken up in saturated NaHCO3Basification to pH 9-10, extraction with EtOAc (200mL x2), combined organic phases over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give compound B108(4.60g) as a brown oil. The crude product was used directly in the next step.
Synthesis procedure for B109
To compound B108(4.60g, crude) was added HCOOH (20mL) and the resulting mixture was stirred at 90 ℃ for 12 h. LCMS showed the reaction progressed well. The reaction was quenched by the addition of 50mL of water and saturated NaHCO3The resulting mixture was extracted with EtOAc (200mL x3), the combined organic phases were washed with brine (150mL) and over anhydrous Na2SO4Drying, filtration and concentration under reduced pressure gave compound B109 as a brown solid (3.70g, 3 steps yield: 83.6%).
Synthesis procedure for B110
Compound B109(2.0g) was followed by the same procedure as B104 to obtain 900mg of compound B110 as a purple powder.
Synthesis procedure for B111
Compound B110(1.2g) was followed by the same procedure as B3 to obtain 1.0g of compound B111 as a white powder.
Scheme 36: synthetic route to B115
Figure BDA0002830360940000741
Synthesis procedure of B113
A mixture of compound B112(1.00g, 5.32mmol) in 1,1, 1-triethoxyethane (4.40g, 27.1mmol) was heated at 120 ℃ and N2Stirring was continued for 2 hours to form a black red solution. LCMS showed 91.7% desired MS. In thatRemoving most of MeC (OEt) under reduced pressure3Compound B113(950mg) was obtained as a red powder.
Synthesis procedure of B114
Compound B113(950mg) was followed by the same procedure as B104 to obtain 230mg of compound B114 as pink powder.
Synthesis procedure for B115
Compound B114(257mg) was followed the same procedure as B3 to obtain 250mg of compound B115 as a yellow gum.
Scheme 37: synthetic route of B122
Figure BDA0002830360940000751
Synthesis procedure for B118
To a stirred solution of compound B116(6.80g, 53.5mmol) in DCM (50mL) at-5-0 ℃ compound B117(8.38g, 48.6mmol) dissolved in DCM (50mL) was added dropwise. The mixture was then stirred at 1-11 ℃ for 64 hours. TLC showed complete consumption of compound B116. The mixture was washed with water (100mL) and brine (100mL), dried over anhydrous sodium sulfate and concentrated to give compound B118(10.0g) as a brown solid.
Synthesis procedure for B119
Mixing compound B118(10.0g, 38.0mmol) and AlCl3(12.7g, 95.2mmol) in N2Stirred at 120 ℃ for 4 hours under an atmosphere. TLC showed complete consumption of compound B90. The mixture was dissolved in DCM (30mL), poured into ice water (50mL) and separated. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the crude product as a brown oil. The crude product was purified by Combi flash chromatography to give compound B119(1.10g) as a yellow solid.
Synthesis procedure for B120
To a stirred solution of compound B119(1.10g, 4.87mmol) in DMSO (30mL) was added KOH (1.09g, 19.4mmol) and MeI (2.07g, 14.5 mmol). The mixture was then stirred at 10 ℃ for 64 hours. LCMS showed complete consumption of compound B119. The mixture was poured into water (100mL) and extracted with EtOAc (50mL x 3). The combined organic layers were washed with brine (50mL x2), dried over anhydrous sodium sulfate and concentrated to give the crude product as a brown oil. The crude product was purified by Combi flash chromatography to give compound B120(840mg) as a yellow oil.
Synthesis procedure for B121
Compound B120(600mg) was followed by the same procedure as B104 to obtain 450mg of compound B121 as a yellow solid.
Synthesis procedure of B122
Compound B121(400mg) was followed by the same procedure as B3 to obtain 180mg of compound B122 as a red oil.
Scheme 38: synthetic route to B125
Figure BDA0002830360940000761
Synthesis procedure for B124
At-10 ℃ and N2To a solution of 2,2,6, 6-tetramethylpiperidine (1.23g, 8.72mmol) in THF (18mL) under an atmosphere was added n-BuLi (2.5M, 2.91mL) for 10 min. A solution of B123(1g, 7.27mmol) in THF (10mL) was then added to the mixture, and the reaction mixture was stirred at-78 deg.C for 10 min. Acetone (844mg, 14.5mmol, 1.07mL) was then added to the reaction mixture. The reaction mixture was stirred at 15 ℃ for 16hr to give a brown mixture. TLC showed a new spot. Subjecting the reaction mixture to NH4Cl (200mL) was quenched and extracted with EA (150 mL. times.2). The organic layer was washed with anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product. The residue was purified by column chromatography to give a pale yellow oilCompound B124(1.3 g).
Synthesis procedure for B125
At 0 ℃ and N2To a solution of B124(1.3g, 6.64mmol) in THF (15mL) under atmosphere was added BH3THF (1M, 33.2mL) and stirred for 30 min. The reaction mixture was then stirred at 60 ℃ for 16 hours to give a colorless mixture. LCMS showed the desired MS and R1 was consumed. The reaction mixture was quenched with MeOH (20mL) and adjusted to pH 2 with 0.5M HCl aq. The mixture was then extracted with DCM (100mL × 2). The aqueous phase was basified to pH 8 with 2M NaOH and extracted with DCM (100mL × 2). The organic layer was washed with anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give B125 as a white solid (800mg, crude).
Scheme 39: synthetic route to B129
Figure BDA0002830360940000771
Synthesis procedure for B127
To the mixture B126(1g, 6.60mmol) in CCl4(10mL) NBS (1.29g, 7.25mmol) and BPO (16mg, 66umol) were added. The reaction mixture was stirred at 85 ℃ for 16hr to give a pale yellow mixture. TLC showed a new spot. The reaction mixture was filtered and the filter cake was washed with CCl4(100mL) washing. The filtrate was concentrated to give a residue. The residue was purified by column chromatography to give B127(1.5g) as a pale yellow solid.
Synthesis procedure for B128
To a solution of B127(1.5g, 6.51mmol) in DMF (15mL) was added NaN3(570mg, 8.77 mmol). The reaction mixture was then heated at 60 ℃ and N2Stirring was carried out under an atmosphere for 16 hours to give a pale yellow mixture. TLC showed a new spot. The reaction mixture was quenched with brine (150mL) and extracted with MTBE (150 mL). The organic layer was washed with brine (100mL) over anhydrous Na2SO4Is dried, filtered and filtered atConcentration under reduced pressure gave B128(1.1g, crude) as a pale yellow oil.
Synthesis procedure for B129
To B128(1.1g, 5.71mmol) in THF (9mL)/H2Solution in O (1mL) Add PPh3(2.25g, 8.57 mmol). The reaction mixture was stirred at 80 ℃ for 16hr to give a brown mixture. LCMS showed the desired MS value. The reaction mixture was diluted with water (100mL) and adjusted to pH 3 with HCl (0.5M), then extracted with EtOAc (100mL x 2). The aqueous phase is treated with NaHCO3The aqueous solution was adjusted to pH 8 and extracted with EtOAc (100 mL). No product was observed by TLC in the organic extracts. The aqueous phase was concentrated to give B129 as a brown solid (2.3g, crude).
Scheme 40: synthetic route to B136
Figure BDA0002830360940000781
Synthesis procedure for B131
Compound B130(100mg) was followed by the same procedure as B41 to obtain 87mg of compound B131 as a brown solid.
Synthesis procedure for B132
Compound B131(87mg) was followed the same procedure as B26 to obtain 90mg of compound B132 as a brown gum.
Synthesis procedure for B133
Compound B132(90mg) was followed by the same procedure as B28 to obtain 80mg of compound B133 as a brown solid.
Scheme 41: synthetic route to B138
Figure BDA0002830360940000782
Synthesis procedure for B135
To SOCl at 0 deg.C2(2.07g, 17.4mmol, 1.26mL) in MeOH (20mL) B134(1g, 8.69mmol) was added. The reaction mixture was stirred at 20 ℃ for 3 hours to give a colorless mixture. TLC showed a new spot. The reaction mixture was concentrated under reduced pressure to give B135(1.3g) as a colorless oil.
Synthesis procedure for B136
The same procedure as for B21 was followed with compound B135(1.3g) to obtain 1.8g of compound B136 as a white solid.
Synthesis procedure of B137
Compound B136(1.8g) was followed by the same procedure as B26 to obtain 1.81g of compound B137 as a yellow gum.
Synthesis procedure for B138
Compound B137(1g) was followed by the same procedure as B125 to obtain 600mg of compound B138 as a yellow gum.
Scheme 42: synthetic route of B143
Figure BDA0002830360940000791
Synthesis procedure of B140
To a solution of compound B139(3g, 11.9mmol) in MeOH (20mL) was added H2SO4(2.38g, 23.8mmol, 1.29 mL). The mixture was refluxed for 17 hours to form a brown solution. TLC showed the reaction was complete. The mixture was dissolved in EtOAc (30mL) and H2Partition between O (30 mL). The aqueous phase was extracted with EtOAc (30mL × 2). The combined organic extracts were washed with brine (50 mL. times.3) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound B140(2.8g) as a yellow solid.
Synthesis procedure for B141
Compound B140(500mg) was followed by the same procedure as B26 to obtain 910mg of compound B141 as a yellow powder.
Synthesis procedure of B142
Compound B141(910mg) was followed by the same procedure as B104 to obtain 560mg of compound B142 as a pale yellow powder.
Synthesis procedure of B143
Compound B142(100mg) was followed by the same procedure as B28 to obtain 100mg of compound B143 as a pale yellow powder.
Scheme 43: synthetic route of B147
Figure BDA0002830360940000801
Synthesis procedure of B146
In N2Next compound B144(792.88mg, 3.06mmol) and compound B145(500mg, 3.06mmol) in dioxane (10mL) and H2Solution in O (2mL) was added Pd (dppf) Cl2(111.95mg, 153.00umol) and Cs2CO3(1.99g, 6.12 mmol). The resulting mixture was heated at 100 ℃ and stirred for 3 hours to give a grey suspension. LCMS showed reaction was not complete. The reaction mixture was then stirred at 100 ℃ for 18 hours and LCMS showed the reaction to have 32% of the desired product and 36% of starting material. The mixture was partitioned between EtOAc (50mL) and water (30 mL). The aqueous layer was extracted with EtOAc (30mL × 3). The combined organic layers were washed with saturated brine (30mL) over anhydrous Na2SO4Dry, filter, and concentrate under reduced pressure to give the crude product as a yellow oil. It was purified by column chromatography to obtain compound B146(236mg) as a yellow oil.
Synthesis procedure of B147
Compound B146(236mg) was followed by the same procedure as a30 to obtain 200mg of compound B147 as a yellow oil.
Scheme 44: synthetic route to Compound 209 via route IThread
Figure BDA0002830360940000811
Synthesis procedure for C2
To a mixture of diisopropylamine (6.69g, 66.2mmol) in dry THF (20mL) at 0 deg.C was added n-BuLi (2.5M, 27.7mL) and stirred at 0 deg.C for 0.5 h, then the mixture was cooled to-70 deg.C and C1(5.00g, 60.2mmol) in THF (20mL) was added to the mixture at-70 deg.C and stirred at-70 deg.C for 0.5 h. The mixture was then heated at-70 ℃ and N2A mixture of ethyl formate (4.90g, 66.2mmol) in THF (20mL) was poured under an atmosphere and the resulting mixture was stirred at-70 ℃ for 0.5 h, then warmed to 15 ℃ and stirred at 15 ℃ for 17 h. TLC (silica gel, PE/EtOAc ═ 2/1) showed the reaction was complete. The reaction mixture was poured into aqueous HCl (150mL, 1M) at 0 ℃ and stirred at 0 ℃ for 0.5 h, then the mixture was extracted with EtOAc (150mL x 3). The organic layer was washed with brine (250mL) over anhydrous Na2SO4The reaction solution was dried and filtered, and the filtrate was concentrated under reduced pressure to give compound C2(5.0g) as a yellow oil. The crude product was used in the next step without further purification.
Synthesis procedure for C3
To a solution of compound C2(8.82g, 67.5mmol) and AcOH (7.09g, 118mmol) in EtOH (5mL) was added NH2-NH2.H2O (4.39g, 87.7mmol), and the resulting mixture was stirred at 78 ℃ for 17 hours to give a pale yellow solution. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give a residue, which was then adjusted to pH 9 with aqueous NaOH (1M), diluted with water (50mL) and extracted with EtOAc (100mL × 3). The organic layer was washed with brine (150mL) over anhydrous Na2SO4Dry above, filter, and concentrate the filtrate under reduced pressure to give compound C3(10.0g, crude) as a yellow gum. The crude product was not further purifiedUsed directly in the next step.
Synthesis procedure for C4
To a mixture of compound C3(5.00g, 40.0mmol) in anhydrous DCM (25mL) at-70 deg.C was added a mixture of ethoxycarbonyl isothiocyanate (4.72g, 36.0mmol) in anhydrous DCM (25mL) and stirred at-70 deg.C for 1 hour, a large amount of white solid appeared. TLC showed the reaction was complete. The mixture was then allowed to warm to-10 ℃ and filtered, and the filter cake was washed with DCM (15mL) to give 4.50g of the desired compound as a white solid, the structure of which was confirmed by HNMR. The filtrate was purified by silica gel column to obtain compound C4(1.80g) as a white solid.
Synthesis procedure for C5
To a mixture of compound C4(6.30g, 24.6mmol) in MeCN (50mL) was added K2CO3(6.79g, 49.2mmol) and the mixture was stirred at 80 ℃ for 8 h. Crude LCMS showed reaction completion. The mixture was cooled to room temperature, then AcOH (15mL) was added to the mixture and stirred at 15 ℃ for 20 min, then the resulting mixture was concentrated under reduced pressure to give a residue which was washed with water (50mL x3) to give compound C5(4.20g) as a white solid.
Synthesis procedure for C6
To a mixture of Compound C5(4.20g, 20.0mmol) in EtOH (40mL) at 15 deg.C was added H2NaOH (2.00g, 50.0mmol) in O (20mL), then MeI (2.84g, 20.0mmol) was added to the above mixture, and the resulting mixture was stirred at 15 ℃ for 2 hours. Crude LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a residue which was treated with ice-cold water (50mL) and aqueous HCl (20mL, 6M) for 30 minutes, giving a large amount of white solid which was filtered to give the crude product. The crude product was poured into MeCN (50mL) to give a suspension, which was then concentrated under reduced pressure to give compound C6(3.60g) as a white solid.
Synthesis procedure for C7
To compound C6(500 m)g, 2.23mmol) in POCl3(5mL) was added N, N-diethylaniline (998mg, 6.69mmol) dropwise. The reaction mixture was stirred at 90 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure to give crude compound C7(710mg) as a dark oil, which was used in the next step without further purification.
Synthesis procedure for C8
To compound B42(700mg, 2.99mmol) in CH3CN (20mL) was added DIEA (772mg, 5.98mmol) and compound C7(652mg, 2.69mmol) and the mixture was stirred at 15 ℃ for 2h to give a yellow mixture. LCMS showed reaction completion. The reaction was concentrated under reduced pressure to give the crude product. The crude product was purified by Combi flash chromatography to give compound C8(650mg) as a yellow gum.
Synthesis procedure for C9
To a mixture of compound C8(650mg, 1.48mmol) in DCM (10mL) was added m-CPBA (659mg, 3.25mmol) in portions at 15 ℃. Reacting the reaction mixture in N2Stirring was carried out at 15 ℃ for 2 hours under an atmosphere to give a yellow mixture. LCMS showed reaction completion. The reaction mixture was filtered, the filtrates combined and concentrated under reduced pressure to give the crude product. The crude product was purified by Combi flash chromatography to give compound C9(350mg) as a yellow gum.
Synthesis procedure for C11
A mixture of compound C9(350mg, 0.740mmol) and compound C10(317mg, 1.48mmol) in NMP (10mL) was stirred at 140 ℃ for 16h to give a brown mixture. LCMS showed reaction completion. The reaction was quenched with water (50mL) and extracted with EtOAc (100mLx 2). The combined organic phases were washed with brine (100 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product, which is purified by Combi flash chromatography to give compound C11(220mg) as a yellow gum.
Synthesis procedure for Compound 209
Will be transformed intoA solution of Compound C11(220mg, 0.362mmol) in HCl/MeOH (3mL, 4M) was stirred at 15 deg.C for 16h to give a yellow solution. LCMS showed reaction completion. The solution was concentrated under reduced pressure to give the crude product. The crude product was purified by preparative HPLC (0.1% TFA) and fractions were purified with saturated NaHCO3Basified to pH 8, extracted with DCM (20mL x2), the separated organic layer was washed with brine (20mL x2) and over anhydrous Na2SO4Dry, filter and freeze-dry to give compound 209(62mg) as a white powder.
Scheme 45: synthesis of compound 245 via scheme I
Figure BDA0002830360940000841
Synthesis procedure for C12
Compound C9(1.46g) and compound a30(680mg) were followed by the same procedure as C11 to obtain 110mg of compound C12 as a yellow solid.
Synthesis procedure for Compound 245
To a mixture of compound C12(110mg, 0.179mmol) in EtOH (5mL) was added N2H4.H2O (12.7mg, 12.3uL, 85% purity). The mixture was stirred at 25 ℃ for 2 hours to give a colorless mixture. LCMS showed reaction completion. The reaction mixture was concentrated under reduced pressure to remove EtOH, and the residue was taken up with H2Dilute O and extract with EtOAc (10mL × 3). The combined organic layers were washed with brine (20mL) over Na2SO4Dry above, filter and concentrate under reduced pressure to give the crude product, which was purified by preparative TLC to give compound 245(17.6mg) as a white powder.
Scheme 46: synthetic route for Compound 48 through route II
Figure BDA0002830360940000851
Synthesis procedure for C13
To a mixture of compound C6(5.00g, 22.3mmol) in DCM (100mL) was added m-CPBA (10.1g, 46.8mmol) portionwise at 25 ℃. Mixing the mixture in N2Stirred under an atmosphere at 25 ℃ for 2 hours. A large amount of white solid appeared. TLC showed the reaction was complete. The reaction mixture was filtered. Most of the DCM was removed under reduced pressure and the mixture was filtered. The filter cake was washed with cold DCM (15mL × 2). This process was repeated twice. The filtrate was washed with anhydrous Na2SO4Dry above then filter and concentrate under reduced pressure to give compound C13(5.7g) as a white powder. The crude product was used in the next step without further purification.
Synthesis procedure for C15
To a solution of compound C13(6.62g, 25.83mmol) in NMP (100mL) was added compound C14(18.15g, 77.5 mmol). The reaction mixture was stirred at 140 ℃ for 16 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between brine (500mL) and EtOAc (400 mL). The organic layer was washed with water (100 mL. times.2), brine (100mL) and washed with anhydrous Na2SO4Dried and concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give a brown gum which was purified with CH3CN (50mL) was triturated to give Compound C15(2.06g) as an off-white powder.
Synthesis procedure for C16
To compound C15(89mg, 0.22mmol) in POCl at 20 deg.C3(4.41g, 28.7mmol) was added N, N-diethylaniline (97mg, 0.65 mmol). The reaction mixture was stirred at 80 ℃ for 2 hours. The reaction mixture was concentrated under reduced pressure to give compound C16(93mg) as a brown gum as a crude product. The crude product was used in the next step without further purification.
Synthesis procedure for C17
To a mixture of compound C16(100mg, 0.233mmol), DIPEA (60.3mg, 0.466mmol) in MeCN (3mL) was added compound B42(109mg, 0.466 mmol). The mixture was stirred at 15 ℃ for 2 hours. TLC showed the reaction was complete. The mixture was filtered. The filter cake was washed with MeCN (1mL x2), PE (1mL x2) to give compound C17(100mg) as a yellow powder.
Synthesis procedure for Compound 48
A mixture of compound C17(90mg, 0.143mmol) in HBr/HOAc (2.5mL, 35% purity) was stirred at 15 ℃ for 20 min. TLC showed a new spot. The mixture was concentrated under reduced pressure to give a residue. Adding saturated NaHCO to the residue3The solution was brought to pH 8. The mixture was extracted with DCM (30mL x 2). The combined extracts were washed with water (40 mL. times.2) over anhydrous Na2SO4Dried, then filtered and concentrated under reduced pressure to give a residue (as a yellow gum). The residue was purified by preparative HPLC and the remaining solvent was removed by freeze-drying to give compound 48(24.3mg) as a white powder.
Scheme 47: synthesis of compound 73 via scheme III
Figure BDA0002830360940000861
Synthesis procedure for C18
To compound E2(300mg, 1.30mmol) in CH3CN (20mL) Compound B42(305mg, 1.30mmol) and K2CO3(180mg, 1.30 mmol). The mixture that could be obtained was heated at 80 ℃ and stirred for 3 hours to obtain a yellow suspension. TLC showed the reaction was complete. The reaction mixture is purified by addition of H2O (50mL) was quenched and extracted with EtOAc (50 mL. times.2). The combined organic layers were washed with brine (20mL) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The crude product was purified by column on silica gel and fractions were concentrated to obtain compound C18(420mg) as a yellow solid.
Synthesis procedure for C20
To a solution of compound C18(200mg, 0.467mmol) in NMP (20mL) was added compound C19(468mg, 2.34 mmol). The resulting mixture was heated at 140 ℃ and stirred for 12hrs to give a yellow solution. TLC showed most of compound 5 was consumed, forming one major spot. The reaction mixture is purified by addition of H2O (100mL) was quenched and extracted with EtOAc (50 mL. times.2). The combined organic layers were washed with brine (10mL) over Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by column of silica gel to obtain compound C20(168mg) as an off-white solid.
Synthesis procedure for Compound 73
To a solution of compound C20(168mg, 0.284mmolq) in DCM (14mL) was added TFA (6 mL). The resulting mixture was stirred at 20 ℃ for 1.5 hours to give a yellow solution. LCMS and HPLC showed the reaction was complete. The reaction mixture was concentrated to remove most of the TFA and then saturated NaHCO was used3The aqueous solution was basified to pH 9 and extracted with EtOAc (50 mL. times.5). The combined organic layers were washed with brine (5 mL. times.5) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by trituration with MTBE (20mL) and filtered under reduced pressure to give compound 73(122.3mg) as an off-white solid.
Exception of synthetic routes
Scheme 48: synthetic route to compound 178
Figure BDA0002830360940000871
Synthesis procedure for D2
To (2-bromophenyl) methylamine (5.14g, 27.7mmol) in CH3CN (50mL) Compound D1(4.00g, 25.1mmol) was added. The mixture was stirred at 85 ℃ for 12 hours,a yellow mixture was obtained. LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a yellow residue which was purified by Combi flash chromatography to give compound D2(3.2g) as a yellow powder.
Synthesis procedure for D3
To a solution of compound D2(1g, 3.23mmol) in DMA (8mL) was added Pd2(dba)3(296mg,0.323mmol)、DPPF(359mg,0.647mmol)、Zn(CN)2(228mg, 1.94mmol), Zn (21.2mg, 0.323 mmol). Reacting at N2Heating at 150 deg.C for 0.5 hr under microwave condition to obtain reddish brown suspension. TLC showed the reaction was complete. The mixture was partitioned between water (50mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (50 mL). The combined extracts were washed with brine (30 mL. times.3) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give the crude product as a brown oil, which was purified by Combi flash chromatography to give compound D3(530mg) as a yellow gum.
Synthesis procedure for D4
To a mixture of Raney-Ni (100mg in water) in MeOH (30mL) was added compound D3(500mg, 1.96mmol), NH3.H2O (2 mL). The suspension is degassed under vacuum and treated with H2And purging for several times. Subjecting the mixture to conditions of 15 ℃ and H2Stirring was carried out for 2 hours (15psi) to give a black suspension. TLC showed the reaction was complete. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give compound D4(500mg) as a yellow powder.
Synthesis procedure for D5
To a mixture of compound D4(500mg, 1.93mmol) in MeCN (5mL) was added DIEA (1mL) and compound C16(827mg, 1.93 mmol). The mixture was stirred at 15 ℃ for 12 hours to give a yellow mixture. TLC showed the reaction was complete. The mixture was partitioned between EtOAc (50mL) and water (50 mL). The aqueous phase was extracted with EtOAc (50mL × 2). The combined extracts were washed with brine (50mL) over Na2SO4Drying on the upper part of the raw materialFiltration and concentration under reduced pressure gave a yellow oil which was purified by Combi flash chromatography to give compound D5(830mg) as a yellow powder.
Synthesis procedure for D6
At-60 ℃ and N2To a mixture of tetrahydro-2H-pyran-4-amine (111mg, 1.10mmol) in DCM (2mL) under an atmosphere was added Al (CH)3)3(2M, 405 uL). The mixture was stirred at 20 ℃ for 1 hour. Compound D5(100mg, 0.162mmol) in anhydrous DCM (1mL) was then added dropwise. The mixture was stirred at 20 ℃ for 12 hours to give a yellow suspension. LCMS showed reaction completion. The reaction mixture was filtered through celite and concentrated under reduced pressure to give a yellow residue which was purified by preparative TLC to give compound D6(53mg) as a yellow gum.
Synthesis procedure for Compound 178
A mixture of compound D6(53.0mg, 0.0750mmol) in HBr/HOAc (1mL, 35% HBr in HOAc) was stirred at 0-10 deg.C for 0.5 h to give a yellow mixture. TLC (PE/EA ═ 1/1) showed the reaction was complete. MTBE (10mL) was added to the reaction mixture to precipitate a red powder. The red powder was collected by filtration and washed with MTBE (5mL x 2). The red solid was purified by passing it through a cation exchange resin using 5% NH3.H2Elution with O, followed by lyophilization gave a white powder which was purified by preparative TLC and lyophilized to give compound 178(20.5mg) as a white powder.
Scheme 49: synthetic route to compound 24
Figure BDA0002830360940000901
Synthesis procedure for D8
Compound D7(50.0g, 430mmol) in SOCl2The mixture in (328g, 2.76mol) was stirred at 78 ℃ for 2 hours to give a pale yellow solution. The solution was concentrated under reduced pressureTo give the acid chloride as a pale yellow oil (65.0g, crude). The crude product was used directly in the next step. To NH3.H2A solution of O (109g, 22%) in DCM (300mL) was added dropwise to the acid chloride (65.0g, crude). The reaction mixture was stirred at 15 ℃ for 0.5 hour. The mixture was extracted with DCM (150mL x3), the combined organic layers were washed with brine (250mL) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound D8(20.5g) as a white powder.
Synthesis procedure for D9
In N2To a solution of compound D8(10.0g, 86.8mmol) in DCM (150mL) under an atmosphere was added TEA (43.9g, 434mmol) and TFAA (45.6g, 217mmol) in turn dropwise. The reaction mixture was stirred at 15 ℃ for 16 hours. Reacting with saturated NH4Quenched with Cl (200mL), then extracted with DCM (100 mL. times.2), and the combined organic layers were washed with brine (150mL) over anhydrous Na2SO4Dry, filter, and concentrate in vacuo to afford compound D9(16.2g, crude) as a yellow oil.
Synthesis procedure for D10
At-70 ℃ and N2To a solution of N-BuLi (2.5M, 56.8mL) in anhydrous THF (200mL) under an atmosphere was added N-isopropylpropan-2-amine (13.7g, 135mmol) dropwise. Mixing the mixture in N2Stirred under an atmosphere at 15 ℃ for 1 hour. Then at-70 ℃ and N2Compound D9(12.0g, 123mmol) in anhydrous THF (20mL) was added to the mixture under atmosphere, and the resulting mixture was heated at-70 ℃ and N2Stirred under atmosphere for 2 hours. At-70 ℃ and N2Dropwise addition of HCO in anhydrous THF (20mL) under atmosphere2Et (12.8g, 173mmol), and the reaction mixture was stirred at 15 ℃ for 16 h. TLC showed a new spot. The reaction was quenched with aqueous HCl (10%), adjusted to pH 3-4, extracted with EtOAc (120mL x2), and the combined organic layers were washed with brine (150mL) and over anhydrous Na2SO4Dry, filter and concentrate in vacuo to give compound D10(13.0g, crude) as a brown oil. What is needed isThe crude product was used in the next step without any purification.
Synthesis procedure for D11
To a solution of compound D10(900mg, 7.19mmol) in EtOH (15mL) was added NH in turn dropwise2NH2·H2O (478mg, 9.35mmol) and AcOH (734mg, 12.2 mmol). The solution was stirred at 78 ℃ for 16 hours. TLC showed a new spot. The solution was concentrated under reduced pressure to give a yellow oil. To the residue were added EtOAc (20mL) and H2O (50mL), the aqueous phase was neutralized with 1M NaOH and the pH was adjusted to 9 and extracted with EtOAc (20 mL. times.2). The combined organic layers were washed with brine (30mL) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound D11(550mg, crude) as a yellow oil, which was used in the next step without any purification.
Synthesis procedure for D12
At-78 ℃ and N2To a solution of compound D11(550mg, 3.95mmol) in DCM (10mL) under an atmosphere was added EtO dropwise2C-NCS (518mg, 3.95 mmol). The reaction solution was stirred at-78 ℃ for 10 minutes. TLC showed the starting material was consumed and some new spots formed. The reaction solution was concentrated under reduced pressure to give a yellow crude oil, which was purified by Combi flash chromatography to give intermediate (230mg) as a yellow solid. To a solution of intermediate (230mg, 0.850mmol) in MeCN (4mL) was added K in one portion2CO3(235mg, 1.70 mmol). The suspension was stirred at 80 ℃ for 1 hour. LCMS showed reaction completion. The suspension was neutralized with AcOH and the pH was adjusted to 4, concentrated under reduced pressure to give a yellow oil. DCM (20mL) was added to dissolve the product, H2O (2 × 20mL) and brine (20 mL). The organic layer was washed with anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound D12(300mg) as a yellow solid.
Synthesis procedure for D13
To compound D12(300mg, 1.34mmol) in EtOH (7mL) and NaOH (2M, 1.34mL)MeI (190mg, 1.34mmol) was added dropwise to the solution in (1). The mixture was stirred at 15 ℃ for 0.5 h. LCMS showed reaction completion. The mixture was concentrated in vacuo to give a residue, DCM (15mL) was added, 6M HCl (2mL) was added and the resulting mixture was stirred for 10 min. The mixture was extracted with DCM (20mL x2), the combined organic layers were washed with water (20mL x2) and brine (30mL) over anhydrous Na2SO4Above was dried, filtered and concentrated in vacuo to give compound D13(160mg) as a yellow solid, which was used in the next step without any purification.
Synthesis procedure for D14
To a solution of compound D13(500mg, 2.10mmol) in N, N-diethylaniline (1.57g, 10.5mmol) was added POCl3(16.5g, 107mmol) and the reaction mixture was stirred at 90 ℃ for 2 hours to give a brown solution. The mixture was concentrated under reduced pressure to remove POCl3. The product was used in the next step without further purification.
Synthesis procedure for D16
To a solution of compound D14(500mg, 1.95mmol) in MeCN (2.00mL) was added DIEA (1.26g, 9.75mmol) followed by compound D15(675mg, 3.90mmol) to the mixture and the resulting mixture was stirred at 20 ℃ for 2 h. LCMS showed the desired MS value. The mixture was poured into water (50mL), extracted with EtOAc (50mL x3), and the combined extracts were washed with brine (100mL) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound D16(524mg) as a white powder.
Synthesis procedure for D17
To compound D16(524mg, 1.33mmol) in CH2Cl2(5.00mL) m-CPBA (573mg, 2.66mmol) was added and the mixture was stirred at 20 ℃ for 1 h. LCMS showed that compound D16 was consumed and the desired MS was obtained. The reaction was saturated with Na2S2O3Aqueous solutionQuench (20mL), extract the mixture with DCM (30 mL. times.3), and combine the extracts with saturated NaHCO3The aqueous solution (30mL) and water (30mL x2) were washed and the organic phase was washed with anhydrous Na2SO4The above was dried, filtered, and the filtrate was concentrated under reduced pressure to give compound D17(450mg) as a yellow oil.
Synthesis procedure for D18
At 120 ℃ and N2Next, to a mixture of compound D17(450mg, 1.06mmol) in NMP (5.00mL) was added compound D5(423mg, 2.12mmol) in one portion. The mixture was stirred at 120 ℃ for 3 hours to give a yellow solution. TLC showed compound D17 was consumed and a new spot was formed. The mixture was poured into water (10mL) and a large amount of white solid appeared, the mixture was filtered, the filter cake was washed with water (10mL) and dried under high vacuum to give compound D18(390mg) as a white powder.
Synthesis procedure for Compound 24
To compound D18(390mg, 0.714mmol) in CH2Cl2(10.0mL) was added TFA (4.84g, 42.4mmol) in one portion. The mixture was stirred at 25 ℃ for 2 hours to give a yellow solution. LCMS showed reaction completion. Adding saturated NaHCO to the reaction solution3The aqueous solution was extracted with DCM (10mL x2) to give pH 7-8 and the combined organic phases were concentrated under reduced pressure to give a brown oil. The crude product was purified by preparative HPLC (0.01% TFA). Most of the MeCN was removed under reduced pressure. The remaining solvent was removed by freeze-drying to give compound 24(161.1mg) as a white powder.
Scheme 50: synthetic route to compound 18
Figure BDA0002830360940000941
Synthesis procedure for D20
To a mixture of compound D19(5.00g, 30.9mmol) in anhydrous DMF (30mL) at 0 deg.C was added ethoxycarbonyl isothiocyanic acid ethyl ester (3.64g, 27.8mmol) and stirred at 0 deg.C for 1 hour, then a large amount of white solid appeared. Crude LCMS showed reaction completion. The mixture was then poured into water (200mL) and a large amount of white solid appeared and the mixture was filtered to give the crude product. The crude product was washed with EtOH (30mL) to give compound D20(4.50g) as a white powder.
Synthesis procedure for D21
To anhydrous EtOH (40mL) was added Na (686mg, 30.0mmol), the mixture was stirred at 20 ℃ for 0.5 h, then Compound D20(3.50g, 11.9mmol) was added to the above mixture, and the resulting mixture was stirred at 80 ℃ for 3 h. Crude LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a residue, which was then adjusted to pH 5 with aqueous HCl (1M) to give a suspension, which was then filtered to give compound D21(2.10g) as a white powder.
Synthesis procedure for D22
To a mixture of compound D21(900mg, 3.64mmol) in EtOH (20mL) was added H2NaOH (364mg, 9.11mmol) in O (10mL), then MeI (517mg, 3.64mmol) was added to the mixture, and the resulting mixture was stirred at 20 ℃ for 17 hours. The two reactions were combined and crude LCMS indicated reaction completion. The mixture was concentrated under reduced pressure to give a residue. The pH of the residue was adjusted to 5 with aqueous HCl (1M), a large amount of white solid appeared, and filtration gave compound D22(1.80g) as a white powder.
Synthesis procedure for D23
To compound D22(800mg, 3.06mmol in POCl)3(6mL) N, N-diethylaniline (2.74g, 18.4mmol) was added and the mixture was stirred at 90 ℃ for 2 h. The mixture was concentrated under reduced pressure to give 4.20g (crude) of compound D23 as a yellow gum.
Synthesis procedure for D24
To chemical combinationA solution of material D23(4.20g, crude) and DIPEA (11.7g, 90.1mmol) in MeCN (10mL) was added compound D15(1.20g, 6.91mmol) and the mixture was stirred at 20 ℃ for 17 h. Crude LCMS showed poor reaction progress and DIPEA (11.7g, 90.1mmol) was then added to the mixture and the mixture was stirred at 20 ℃ for 17 hours. Crude LCMS showed reaction completion. The mixture was poured into water (50mL), extracted with EtOAc (50mL x3), and the combined extracts were washed with brine (100mL) over anhydrous Na2SO4Dry above, filter and concentrate the filtrate under reduced pressure to give a residue which is purified by Combi flash chromatography to give 300mg of compound D24 as a yellow gum.
Synthesis procedure for D25
To a solution of compound D24(300mg, 0.721mmol) in DCM (10mL) was added m-CPBA (326mg, 1.51mmol, 80% purity) and the mixture was stirred at 20 ℃ for 1 h. A large amount of white solid appeared. Crude LCMS showed reaction completion. The reaction was saturated with Na2S2O3The aqueous solution (3mL) was quenched, then the mixture was poured into DCM (100mL) and saturated NaHCO3The aqueous solution (100mL x3) and brine (100mL x2) were washed and the organic phase was purified over anhydrous Na2SO4The above was dried, filtered, and the filtrate was concentrated under reduced pressure to give 250mg of compound D25 as a white powder.
Synthesis procedure for D26
To a solution of compound D25(250mg, 0.558mmol) in NMP (3mL) was added compound D5(223mg, 1.12mmol), and the mixture was heated at 120 ℃ and N2Stirred under atmosphere for 1 hour. Crude LCMS showed reaction completion. After cooling to room temperature, the mixture was poured into water (30mL) and a large amount of white solid appeared, the mixture was filtered and the filter cake was washed with water (50mL) to give 153mg of compound D26 as a yellow powder.
Synthesis procedure for Compound 18
To a solution of compound D26(150mg, 0.264mmol) in DCM (3mL) was added TFA (4.62g, 40.5mmol) and the mixture wasThe mixture was stirred at 20 ℃ for 20 minutes. Crude LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a residue, which was taken up in saturated NaHCO at pH3The aqueous solution was adjusted to 8, diluted with water (20mL), the mixture was extracted with DCM (30mL x3), and the combined extracts were washed with brine (50mL) over anhydrous Na2SO4Dry above, filter and concentrate the filtrate under reduced pressure to give a residue which is purified by preparative HPLC (0.05%, HCl salt). Removal of most of the CH by evaporation under reduced pressure3CN, and the remaining solvent was removed by freeze-drying to give 78.6mg of compound 18 as a white powder.
Scheme 51: synthetic route to compound 35
Figure BDA0002830360940000961
Synthesis procedure for D28
At 120 ℃ and N2To a mixture of compound D26(500mg, 0.879mmol) in dioxane (5mL) under atmosphere was added compound D27(952mg, 2.64mmol) and Pd (PPh) in one portion3)4(101mg, 0.0879 mmol). The mixture was stirred at 120 ℃ for 16h to give a yellow solution. LCMS showed 10.4% desired MS. Pouring the mixture over NH4Aqueous Cl (50mL) and stirred for 10 min. The mixture was partitioned between EtOAc (50mL) and water (50 mL). The aqueous phase was extracted with EtOAc (50 mL). The combined organic extracts were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The crude product was purified by Combi flash chromatography to give compound D28(91.6mg) as a white powder.
Synthesis procedure for Compound 35
To compound D28(110mg, 0.206mmol) in CH2Cl2(2mL) was added TFA (1.40g, 12.3mmol) in one portion. Stirring the mixture at 25 ℃ for 1 hour to obtainTo a yellow solution. LCMS showed the reaction was complete and showed 52.8% desired MS. Adding saturated NaHCO to the reaction solution3The aqueous solution was adjusted to pH 7-8, extracted with DCM (10mL x2), and the combined organic phases were concentrated under reduced pressure to give a brown oil which was purified by preparative HPLC (0.01% TFA). Most of the MeCN was removed under reduced pressure. The remaining solvent was removed by freeze-drying to give compound 35(10.40mg) as a white powder.
Scheme 52: synthetic route to compound 30
Figure BDA0002830360940000971
Synthesis procedure for D29
To compound D26(50mg, 0.088mmol), 1-pentenylboronic acid (50mg, 0.44mmol), Na2CO3(19mg, 0.18mmol) in H2Mixture of O (0.5mL) and dioxane (2mL) with Pd (dppf) Cl2(13mg, 0.018mmol), the mixture was washed with N2Purging once, then putting the mixture in N2Stirred under an atmosphere of microwaves at 110 ℃ for 1 hour. Crude LCMS showed the desired MS value. The mixture was poured into water (30mL), extracted with DCM (30mL × 3), and the combined extracts were washed with brine (50mL) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to yield 150mg of compound D29 as a yellow gum.
Synthesis procedure for D30
To a solution of compound D29(120mg, 0.215mmol) in MeOH (10mL) was added Pd/C (20mg) and the mixture was taken up with H2Purging three times and H at 20 ℃2Stir under balloon (15psi) for 3 hours. Crude LCMS showed reaction completion. The mixture was filtered and the filtrate was concentrated under reduced pressure to give 100mg of compound D30 as a yellow gum.
Compound (I)Synthesis procedure of 30
To a solution of compound D30(100mg, 0.179mmol) in DCM (3mL) was added TFA (2mL) and the mixture was stirred at 20 ℃ for 1 h. Crude LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a residue, which was taken up in saturated NaHCO at pH3The aqueous solution was adjusted to 8, extracted with DCM (30 mL. times.3) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (0.05% HCl). Removal of most of the CH by evaporation under reduced pressure3CN, and the remaining solvent was removed by freeze-drying to obtain 10.5mg of compound 30 as a white powder.
Scheme 53: general synthetic route to compound 163
Figure BDA0002830360940000981
Synthesis procedure for D33
To a solution of compound D31(1.00g, 2.43mmol) in dry THF (10mL) at 0 deg.C was added compound D32(1.28g, 6.08mmol) and the mixture was stirred at 0 deg.C for 2 h. Crude LCMS showed reaction completion. The reaction was quenched with brine (100mL), the mixture was extracted with EtOAc (100mL x2), and the combined extracts were washed with brine (100mL) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to yield 600mg of compound D33 as a yellow gum.
Synthesis procedure for D34
To a solution of compound D33(550mg, 1.25mmol) in DCM (3mL) at 0 deg.C was added BBr3(627mg, 2.50mmol) and the mixture was then allowed to warm to 20 ℃ and stirred at 20 ℃ for 3 hours. Crude LCMS showed the reaction progressed well. Reacting with saturated NH4Aqueous Cl (30mL) and the pH of the mixture was adjusted withSaturated NaHCO3The aqueous solution was adjusted to 8, extracted with EtOAc (50 mL. times.3), and the combined extracts were washed with brine (100mL) over anhydrous Na2SO4The mixture was dried, filtered and the filtrate was concentrated under reduced pressure to give 600mg of crude product, which was confirmed by HNMR. 100mg was used in the next step, but there was no reaction. The crude product was then purified by Combi flash chromatography to yield 360mg of compound D34 as a yellow gum.
Synthesis procedure for D36
To compound D34(310mg, 0.729mmol), compound D35(409mg, 2.19mmol) and PPh at 0 deg.C3(382mg, 1.46mmol) of DEAD (254mg, 1.46mmol) was added to a mixture in dry THF (2mL), then the mixture was allowed to warm to 20 ℃ and stirred at 20 ℃ for 17 h. Crude LCMS showed the reaction progressed well. The reaction was quenched with water (30mL), extracted with EtOAc (30mL x3), and the combined extracts were washed with brine (50mL) over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to yield 150mg of compound D36 as a yellow gum.
Synthesis procedure for Compound 163
To a solution of compound D36(150mg, 0.252mmol) in DCM (3mL) was added TFA (3mL) and the mixture was stirred at 20 ℃ for 0.5 h. Crude LCMS showed reaction completion. The mixture was concentrated under reduced pressure to give a residue. The pH of the mixture is then adjusted with NaHCO3The aqueous solution was adjusted to pH 8, then diluted with water (15mL), extracted with DCM (30mL x3), and the combined extracts were washed with brine (50mL) and over anhydrous Na2SO4The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (0.05% HCl). Removal of most of the CH by evaporation under reduced pressure3CN, and the remaining solvent was removed by freeze-drying to give 27.8mg of compound 163 as a yellow gum.
Scheme 54: synthetic route to compound 23
Figure BDA0002830360940001001
Synthesis procedure for D37
To a solution of compound D31(1.00g, 2.43mmol) in dioxane (10mL) was added NaOH (800mg, 20.0mmol) and H2O (10mL), and the mixture was stirred at 50 ℃ for 16 hours to give a yellow solution. TLC showed the reaction was complete. The reaction solution was concentrated under reduced pressure to remove dioxane, and then 6M HCl was added dropwise to the reaction mixture until pH 5. The mixture was extracted with DCM (20mL × 2) over anhydrous Na2SO4Dry above, filter, and concentrate under reduced pressure to give compound D37(952mg) as a yellow solid.
Synthesis procedure for D38
To compound D37(952mg, 2.72mmol) in POCl3(10mL) N, N-diethylaniline (1.2g, 8.16mmol) was added and the reaction mixture was stirred at 90 ℃ for 2 hours to give a brown solution. LCMS showed reaction completion. The mixture was concentrated under reduced pressure to remove POCl3The residue was then purified in DCM (20mL) and saturated NaHCO3Partitioned (20mL), the aqueous phase extracted with DCM (20 mL. times.2), the combined organic phases washed with brine (40mL) over anhydrous Na2SO4Dry, filter, and concentrate under reduced pressure to give a brown oil. The crude product was purified by Combi flash chromatography to give compound D38(828mg) as a yellow oil.
Synthesis procedure for D40
To a solution of compound D38(200mg, 0.544mmol) and compound D39(202mg, 0.652mmol) in dioxane (4mL) was added H2K in O (1mL)2CO3(188mg, 1.36mmol) and Pd (dppf) Cl2(20mg, 0.272mmol) and the mixture was stirred under N2Balloon stirring was carried out at 90 ℃ for 16 hours to obtain a black solution. LCMS showed 43.8% of desired MS value. To the solutionAddition of H2O (10mL), extracted with EtOAc (10mL x2), and the combined organic phases washed with brine (10mL) over anhydrous Na2SO4Dry, filter, and concentrate under reduced pressure to give a brown oil, which was purified by Combi flash chromatography to give compound D40(119mg) as a yellow oil.
Synthesis procedure for D41
To a suspension of compound D40(181mg, 0.352mmol) in MeOH (5mL) was added Pd/C (50% moisture, 10% Pd) and the mixture was taken up in H2Stirring was carried out under a balloon (15psi) at 25 ℃ for 16 hours to give a black suspension. LCMS showed reaction completion. The suspension was filtered through a pad of celite and the combined filtrates were concentrated under reduced pressure to give compound D41(121mg) as a yellow oil.
Synthesis procedure for Compound 23
To a solution of compound D41(121mg, 0.234mmol) in DCM (4mL) was added TFA (1mL) and the reaction solution was stirred at 25 ℃ for 2h to give a yellow solution. LCMS showed reaction completion. Adding saturated NaHCO to the reaction solution3Aqueous solution (10mL), extracted with DCM (10mL × 2) and the combined organic phases concentrated under reduced pressure to give a brown oil. The crude product was purified by preparative HPLC (0.05% HCl). Adding saturated NaHCO to the eluate containing the desired product3Until pH 7, extract with DCM (20mL x3) and concentrate the combined organic phases under reduced pressure. The remaining aqueous solution was lyophilized to give compound 23(3.9mg) as a white powder.
Scheme 55: synthetic route to compound 165
Figure BDA0002830360940001021
Synthesis procedure for D43
To compound D38(120mg, 0.326mmol), compound D42(235mg, crude, ca. 0.489mmol), Pd (PPh)3)4(65mg,0.056mmol) in dioxane (3mL) and H2Na was added to a solution of O (750uL)2CO3(86mg, 0.82 mmol). Reacting the reaction mixture in N2Next, the mixture was stirred at 110 ℃ for 1 hour under microwave conditions. TLC (PE/EtOAc: 5/1, SiO2) Indicating that the reaction was complete. The reaction mixture was diluted with DCM (10mL) and water (10mL) and then filtered. The filtrate was separated. The organic layer was washed with anhydrous Na2SO4Dry, filter, and concentrate under reduced pressure to give the crude product as a brown gum, which was purified by Combi flash chromatography to give compound D43(126mg) as a colorless oil. The impure product was used in the next step without further purification.
Synthesis procedure for Compound 165
To a solution of compound D43(133mg, crude) in DCM (1.6mL) was added TFA (400 uL). The reaction solution was stirred at 25 ℃ for 1 hour. LCMS showed reaction completion. The reaction solution was diluted with DCM (10mL) and water (5 mL). To the mixture was added aqueous ammonia (0.5mL, 28%) until the aqueous layer pH > 7. The organic layer was separated and washed with water (10mL) then concentrated under reduced pressure to give the crude product as a brown oil which was purified by preparative HPLC (0.05% HCl as additive). Most of the MeCN was removed under reduced pressure and the remaining solvent was removed by freeze-drying to give compound 165(35.9mg) as an off-white powder.
Scheme 56: synthetic route to compound 171
Figure BDA0002830360940001031
Synthesis procedure for D45
To compound D38(200mg, 0.54mmol) and K2CO3(187mg, 1.36mmol) in dioxane (1mL) and H2Solution in O (0.5mL) was added Compound D44(176mg, 0.979mmol) and Pd (dppf) Cl2(39.7mg, 0.54 mmol). Reacting the reaction mixture in N2Stirred under an atmosphere at 110 ℃ for 16 hours. L isCMS showed the desired MS value of 82.4%. The mixture was dissolved in EtOAc (100mL) and H2Partition between O (100 mL). The aqueous layer was extracted with EtOAc (100 mL). The combined organic extracts were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry above, filter and concentrate under reduced pressure to give compound D45 as a red powder.
Synthesis procedure for D46
In N2To a solution of compound D45(100mg, 0.221mmol) and compound D5(53mg, 0.26mmol) in anhydrous DMF (1mL) under atmosphere was added HATU (104mg, 0.276mmol) and TEA (37mg, 0.36 mmol). Subjecting the mixture to conditions of 25 ℃ and N2Stirred under atmosphere for 16 hours. LCMS showed the desired MS value of 74.3%. The mixture was dissolved in EtOAc (50mL) and H2Partition between O (50 mL). The aqueous layer was extracted with EtOAc (50 mL). The combined organic extracts were washed with water (50 mL. times.2) over anhydrous Na2SO4Dry, filter and concentrate under reduced pressure to give a residue. The residue was purified by Combi flash chromatography to give compound D46(95.0mg) as a yellow oil.
Synthesis procedure for Compound 171
To compound D46(95mg, 0.15mmol) in CH2Cl2(4mL) was added TFA (1mL) in one portion. The mixture was stirred at 25 ℃ for 1 hour to give a yellow solution. LCMS showed reaction completion and desired MS value of 98.7%. Adding saturated NaHCO to the reaction solution3The aqueous solution was adjusted to pH 7-8, extracted with DCM (10mL x2), and the combined organic phases were concentrated under reduced pressure to give a yellow oil which was purified by preparative HPLC (0.05% HCl). Most of the MeCN was removed under reduced pressure. The remaining solvent was removed by freeze-drying to give compound 171(41.8mg) as a yellow powder.
Reference to the literature
Bregman, r.g.pest and v.j.kidd, cell cycle regulation and RNA polymerase ii (cell cycle regulation and RNA polymerase ii), Front biosci.2000 feb; 1(5) D244-57.
D.desai, h.c.wessling, r.p.fisher and d.o.morgan, the effect of phosphorylation by CAK on binding of cyclin by CDC2 and CDK2 (Effects of phosphorylation by CAK on cyclin binding by CDC2 and CDK2), mol.cell biol.5jan; 15(1):345-350.
Akhtar, M.Heidemann, J.R.Tietjen, D.W.Zhang, R.D.Chapman, D.Eick and A.Z.Ansari, TFIIH Kinase Places a Bivalent tag on the Carboxy-Terminal Domain of RNA Polymerase II (TFIIH Kinase plant Bivalent Marks on the carbon-Terminal Domain of RNA Polymerase II), mol.cell.2009May; 15; 34(3):387-93.
Larochelle, r.amat, k.g.cutter, m.sans unit, c.zhang, j.j.allen, k.m.shokat, d.l.bentley and r.p.fisher, Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II, nat.struct.mol.biol.2012nov; 19(11):1108-15.
Shapiro, Cyclin-Dependent Kinase pathway as a target for Cancer therapy (Cyclin-Dependent Kinase as Targets for Cancer Treatment), j.clin.oncol.2006 apr; 10; 24(11):1770-83.
Locli and l.n.johnson, CAK-cyclin dependent activated kinase: key kinases in cell cycle control and targets for drugs? (CAK-cycle-dependent Activating Kinase: a key Kinase in Cell cycle control and a target for drugs; 4(4):572-7.
Lee and r.a.young, Transcriptional Regulation and its misregulation in Disease (Transcriptional Regulation and its mistreatment in Disease), cell.2013mar; 14; 152(6):1237-51.
S.Nekhai,M.Zhou,A.Fernandez,W.S.Lane,Ned J.C.Lamb,J.Brady,A.Kumar.Biochem.J.2002Jun;15;364(Pt 3):649-57.
Y.k.kim, c.f.bourgeois, r.pearson, m.tyagi, m.j.west, j.wong, s.y.wu, c.m.chiang and j.karn, the Recruitment of TFIIH to the HIV LTR is the rate-limiting step in the expression of HIV from latency (recombination of TFIIH to the HIV LTR is a rate-limiting step in the expression of HIV from latency), embo.j.2006 aug; 9; 25(15):3596-3604.
Kapasi and d.h.spector, the inhibitory specificity of Cyclin-Dependent Kinases at the onset of Human Cytomegalovirus Infection Alters the level and Localization of RNA Polymerase II carboxy-Terminal Domain Kinases cdk9 and cdk7 on Viral transcripts (Inhibition of the cycle-Dependent Kinases at the Beginning of the beginnings of Human Cytomegalovirus Infection of Human cytokine Infection specificity the Levels and Localization of the RNA Polymerase II carboxy-Terminal Domain Kinases cd 9 and cdk7 at the Viral transcriptoposome), j.virol.2008n; 82(1):394-407.
Eickhoff et al, Pyrazolo-triazine derivatives as selective cyclin-dependent kinase inhibitors (Pyrazolo-triazine derivitives) PCT WO2013/128028A1
TABLE 1 enzymatic Activity of CDK (1, 2, 5 and 7) and selectivity of CDK7
Figure BDA0002830360940001061
Figure BDA0002830360940001071
TABLE 1 continuation
Figure BDA0002830360940001072
Figure BDA0002830360940001081
TABLE 1 continuation
Figure BDA0002830360940001082
Figure BDA0002830360940001091
TABLE 1 continuation
Figure BDA0002830360940001101
Figure BDA0002830360940001111
TABLE 1 continuation
Figure BDA0002830360940001112
Figure BDA0002830360940001121
TABLE 1 continuation
Figure BDA0002830360940001122
Figure BDA0002830360940001131
TABLE 1 continuation
Figure BDA0002830360940001141
Figure BDA0002830360940001151
TABLE 1 continuation
Figure BDA0002830360940001152
Figure BDA0002830360940001161
TABLE 1 continuation
Figure BDA0002830360940001162
Figure BDA0002830360940001171
TABLE 2 HCT116 viability assay
Figure BDA0002830360940001172
Figure BDA0002830360940001181
TABLE 3H 460 viability assay
Figure BDA0002830360940001182
Figure BDA0002830360940001191
TABLE 3 continuation
Figure BDA0002830360940001192
Figure BDA0002830360940001201
TABLE 4 MM.1S viability assay
Figure BDA0002830360940001202
Figure BDA0002830360940001211
TABLE 5 MV4-11 viability assay
Figure BDA0002830360940001212
Figure BDA0002830360940001221
TABLE 5 continuation
Figure BDA0002830360940001231
TABLE 6 MOLT4 viability assay
Figure BDA0002830360940001232
Figure BDA0002830360940001241
TABLE 7 viability assay of RPMI-8226
Figure BDA0002830360940001242
Figure BDA0002830360940001251
TABLE 8A 2780 viability assay
Figure BDA0002830360940001252
Figure BDA0002830360940001261
TABLE 9 OVCAR-3 viability assay
Figure BDA0002830360940001262
TABLE 10 comparative data on CDK7 Selectivity profiles in the CDK family
Figure BDA0002830360940001263
Figure BDA0002830360940001271
TABLE 11. Compounds 1-279 summarized in terms of structure and corresponding features
Figure BDA0002830360940001272
Figure BDA0002830360940001281
Figure BDA0002830360940001291
Figure BDA0002830360940001301
Figure BDA0002830360940001311
Figure BDA0002830360940001321
Figure BDA0002830360940001331
Figure BDA0002830360940001341
Figure BDA0002830360940001351
Figure BDA0002830360940001361
Figure BDA0002830360940001371
Figure BDA0002830360940001381
Figure BDA0002830360940001391
Figure BDA0002830360940001401
Figure BDA0002830360940001411
Figure BDA0002830360940001421
Figure BDA0002830360940001431
Figure BDA0002830360940001441
Figure BDA0002830360940001451
Figure BDA0002830360940001461
Figure BDA0002830360940001471
Figure BDA0002830360940001481
Figure BDA0002830360940001491
Figure BDA0002830360940001501
Figure BDA0002830360940001511
Figure BDA0002830360940001521
Figure BDA0002830360940001531
Figure BDA0002830360940001541
Figure BDA0002830360940001551
Figure BDA0002830360940001561
Figure BDA0002830360940001571
Figure BDA0002830360940001581
Figure BDA0002830360940001591
Figure BDA0002830360940001601
Figure BDA0002830360940001611
Figure BDA0002830360940001621
Figure BDA0002830360940001631
Figure BDA0002830360940001641
Figure BDA0002830360940001651
Figure BDA0002830360940001661
Figure BDA0002830360940001671
Figure BDA0002830360940001681
Figure BDA0002830360940001691
Figure BDA0002830360940001701
Figure BDA0002830360940001711
Figure BDA0002830360940001721
Figure BDA0002830360940001731
Figure BDA0002830360940001741
Figure BDA0002830360940001751
Figure BDA0002830360940001761
Figure BDA0002830360940001771
Figure BDA0002830360940001781
Figure BDA0002830360940001791
Figure BDA0002830360940001801
Figure BDA0002830360940001811
Figure BDA0002830360940001821
Figure BDA0002830360940001831
Figure BDA0002830360940001841
Figure BDA0002830360940001851
Figure BDA0002830360940001861
Figure BDA0002830360940001871
Figure BDA0002830360940001881
Figure BDA0002830360940001891
Figure BDA0002830360940001901
Figure BDA0002830360940001911
Figure BDA0002830360940001921
Figure BDA0002830360940001931
Figure BDA0002830360940001941
Figure BDA0002830360940001951
Figure BDA0002830360940001961
Figure BDA0002830360940001971
Figure BDA0002830360940001981
Figure BDA0002830360940001991
Figure BDA0002830360940002001

Claims (19)

1. A compound having the general formula I
Figure FDA0002830360930000011
Wherein
X is independently at each occurrence selected from CH and N;
L1is absent or independently selected at each occurrence from the group consisting of-NH-, -NH (CH)2)-、-NH(C=O)-、-NHSO2-、-O-、-O(CH2) -, - (C ═ O) NH-, and- (C ═ O) (CH)2) -a group of compositions;
q is independently at each occurrence selected from the group consisting of C3-C8 cycloalkyl, aryl, heteroaryl, heterocyclyl, and C1-C6 alkyl, wherein C1-C6 alkyl is-OR5、–N(R5)R5Aryl, heteroaryl and heterocyclyl, substituted with one or both,
C3-C8 cycloalkyl may be substituted by R3、R4And- (C ═ O) R5One or both of them may be substituted,
the heterocyclic radical may be substituted by R3、R4And- (C ═ O) R5One or both of them may be substituted,
the aryl OR heteroaryl group may be substituted by C1-C6 alkyl, -OR5、-N(R5)R5、-(C=O)R5One or both of halo, heteroaryl and heterocyclyl;
R1independently at each occurrence selected from the group consisting of hydrogen and methyl;
R2independently at each occurrence, is selected from the group consisting of halogen, C1-C6 alkyl, C3-C10 cycloalkyl, -CN, - (C ═ O) CH3、-NR9R12And C1-C3 haloalkyl, any of which is optionally substituted;
R3independently at each occurrence, is selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R4independently at each occurrence, is selected from hydrogen, halogen, -OR5、-N(R5)R5Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R5independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C3 haloalkyl, heteroaryl, heterocyclyl, substituted with halogen, -OR11、-N(R11)R11C1-C6 alkyl and optionally-OH or-NH2Heteroaryl substituted by one OR both of substituted C1-C6 alkyl, by halogen, -OR11、-N(R11)R11C1-C6 alkyl and optionally-OH or-NH2A heterocyclic group substituted with one or both of substituted C1-C6 alkyl;
z is any structure in the following group A;
Figure FDA0002830360930000021
wherein
X1At each occurrenceIndependently selected from CR24And N;
X2independently at each occurrence is selected from CR25And N;
R6independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkyl substituted with-OH, C3-C10 cycloalkyl, C3-C10 heterocyclyl, - (C ═ O) NHR11,-NHR9,-NH(C=O)NHR11,-N(CH3)(C=O)CH3,-NH(C=O)R12,-NR9R12,-OR12And any structure of the following group B;
Figure FDA0002830360930000031
R7independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, -NH (C ═ O) R12、-NR9R12、-OR12And any structure from group C below;
Figure FDA0002830360930000032
R8and R10Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2、-OH、-OR5、-CN、-(C=O)R5、-(C=O)OR5、-(C=O)NH2、-(C=O)NHR21、-CH2(C=O)NHR21、-NH(C=O)R13、-NHS(=O)2R5、-S(=O)2NH2、-S(=O)2NHR21And by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R9independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OR5-CN, -C3-C10 cycloalkyl, -C3-C10 heterocyclyl and a substituted group-OH OR-OR5Substituted C1-C6 alkyl;
R11independently at each occurrence, is selected from the group consisting of hydrogen, C1-C6 alkyl, and C3-C10 cycloalkyl;
R12absent OR independently at each occurrence selected from the group consisting of C1-C6 alkyl, substituted by-OR5or-N (R)5)R5Substituted C1-C6 alkyl, C6-C10 aryl, phenyl, benzyl, C3-C9 heteroaryl, C3-C6 heterocyclyl, benzyl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C9 heteroaryl substituted with 1 to 4 halogens or C1-C3 alkyl, C3-C6 heterocyclyl substituted with C1-C3 alkyl, and R (C ═ O) R with 1 to 4 halogens and/or 1 to 4-NH (C ═ O)13Substituted C6-C10 aryl;
R13independently at each occurrence is selected from hydrogen, C1-C6 alkyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl and C3-C10 cycloalkyl;
R14and R15Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, substituted with-OH or-NH2Substituted C1-C6 alkyl, C3-C10 cycloalkyl, - (C ═ O) R5,-(C=O)NHR21,-C(R9)(R11)OR21,-NH(C=O)R21,-NR9R21,-OR21,-OC(R9)(R11)(R21) C3-C10 heterocyclyl, by R4Substituted C3-C10 heterocyclyl, C3-C10 heteroaryl substituted by 1 to 4 halogens or C1-C3 alkyl, C6-C10 aryl such as phenyl and substituted by- (C ═ O) R5、-(C=O)OR5、-(C=O)NH2、-(C=O)NHR21、-CH2(C=O)NHR21、-NH(C=O)R13、-NHS(=O)2R5、-S(=O)2NH2or-S (═ O)2NHR21Substituted aryl groups;
R16independently at each occurrence, is selected from hydrogen, C1-C6 alkyl, - (C ═ O) R13And is-OR5Substituted C1-C6 alkyl;
R17、R18、R19and R20Independently at each occurrenceIs selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, C6-C10 aryl such as phenyl, -CN, -CHCF3NR9R11,-OH,-OR21,-NO2,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-NH(C=O)R13,-NHR5,-NHS(=O)2R5,-S(=O)2NH2,-S(=O)2NHR21And is substituted by-CN, -OH, -OR5、-(C=O)NHR5、-NH2、-NH(C=O)R5、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl;
R21independently at each occurrence, is selected from the group consisting of C1-C6 alkyl, C3-C10 cycloalkyl, C3-C10 heterocyclyl, C1-C3 haloalkyl, aryl, phenyl, benzyl, substituted with-CN, -OH, -OR5、-NH2、-NHR5or-N (R)5)R5Substituted C1-C6 alkyl, aryl substituted by halogen or C1-C3 haloalkyl, C3-C10 heteroaryl substituted by 1 to 4 halogen or C1-C3 alkyl and R4Substituted C3-C10 heterocyclyl;
R22and R23Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OH, -OR5-CN and by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R24and R25Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -NH2,-OH,-OR5,-CN,-(C=O)R5,-(C=O)OR5,-(C=O)NH2,-(C=O)NHR21,-CH2(C=O)NHR21,-NH(C=O)R13、-NHS(=O)2R5,-S(=O)2NH2or-S (═ O)2NHR21And by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
provided that when Z is
Figure FDA0002830360930000051
When then R is6And R7One is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not CH3Cl or 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not phenyl, CH (CH)3)2、CH2CH3Or CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is F, R7Is H, R8Is H and R9Is H, then R10Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not F;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R4Is H, X is N, Z is phenyl, R6Is 1H-pyrazole, R7Is H, R8Is H, R9Is H and R10Is H, then R3Is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is H, R8Is H, R9Is H and R10Is H, then R7Is not Cl;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted heterocyclyl radical, R3Is CH3,R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is absent, Q is substituted by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q is by R3And R4Substituted C3-C8 cycloalkyl, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein,if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is H, R4Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q is represented by R3And R4Substituted heterocyclyl radical, R3Is N (R)5)R5,R4Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Or enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates or pharmaceutically acceptable salts thereof of the above compounds.
2. A compound according to claim 1, which is a pharmaceutically acceptable salt thereof,
wherein R is1Is hydrogen and said compound has the general formula II
Figure FDA0002830360930000091
X, Q, L therein1、R2And Z is as defined in claim 1.
3. The compound of claim 1, having the general formula III
Figure FDA0002830360930000092
X, L therein1、R1、R2And Z is as defined in claim 1, and
Q1absent OR independently at each occurrence selected from the group consisting of aryl, heteroaryl, heterocyclyl, substituted with C1-C6 alkyl, -OR5、-N(R5)R5And aryl substituted by one OR both of halogen, C1-C6 alkyl, -OR5、-N(R5)R5And heteroaryl substituted with one or both of halogen and R29And R30A heterocyclic group substituted with one or both of them;
R29absent OR independently at each occurrence selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
R30independently at each occurrence, is selected from hydrogen, halogen, -OR5、-N(R5)R5Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
wherein R is5、R9And R12As defined in claim 1;
L2absent or independently at each occurrence selected from the group consisting of-O-, -NH-, - (C ═ O) -, and- (C ═ O) NH-;
Y1independently at each occurrence, selected from CH, C (OH), and N;
Y2independently at each occurrence, selected from CH, CR30O and N;
m is independently selected at each occurrence from 0, 1 and 2;
n is independently selected at each occurrence from 0 and 1;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not CH3Cl or 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not phenyl, CH (CH)3)2、CH2CH3Or CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not Cl;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is Cl, R7Is H, R8Is H and R9Is H, then R10Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is Cl, then R6Is not CH3
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is F, R7Is H, R8Is H and R9Is H, then R10Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not F;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is N, m is 1, N is 1, R30Is H, XIs N, Z is phenyl, R6Is 1H-pyrazole, R7Is H, R8Is H, R9Is H and R10Is H, then R29Is not H;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH (CH)3)2
Wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1N is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 2, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 2, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is F, then R6Is not 1H-pyrazole;
wherein if R is1Is H, R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 2, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R7Is H, R8Is H, R9Is H and R10Is H, then R6Is not 1H-pyrazole;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is H, R8Is H, R9Is H and R10Is H, then R7Is not Cl;
wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1, N is 1, R29Is CH3,R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is O, Q1Is absent, L2Is absent, Y1Is CH, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH, Y2Is N, m is 1N is 0, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is OR12,R7Is H, R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is NH, Q1Is absent, L2Is absent, Y1Is CH,Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is N, m is 1, N is 1, R29Is H, R30Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Is- (C ═ O) -, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 1, n is 1, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Wherein if R is1Is CH3,R2Is CH (CH)3)2,L1Absent, Q1Is absent, L2Is absent, Y1Is N, Y2Is CH, m is 2, n is 0, R29Is N (R)5)R5,R30Is H, R5Is H, X is N, Z is phenyl, R6Is H, R7Is OR12,R8Is H, R9Is H and R10Is H, then R12Is not CH3
Or enantiomers, stereoisomeric forms, mixtures of enantiomers, diastereomers, mixtures of diastereomers, racemates or pharmaceutically acceptable salts thereof of the above compounds.
4. A compound according to any one of claims 1 to 3, having general formula IV
Figure FDA0002830360930000151
X, L therein1And R2As defined in claim 1;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined in claim 3;
wherein Z1Is any one of the following group D;
Figure FDA0002830360930000152
wherein R is6、R7、R8、R9And R10As defined in claim 1.
5. The compound of any one of claims 1-4, having general formula V
Figure FDA0002830360930000161
X, X therein1、X2、L1、R2、R6、R22And R23As defined in claim 1;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined in claim 3.
6. The compound of any one of claims 1-4, having general formula VI
Figure FDA0002830360930000162
X, L therein1And R2As defined in claim 1;
wherein m, n, Y1、Y2、L2、R29、R30And Q1As defined in claim 3;
X3independently at each occurrence is selected from CR10And N;
R26、R27and R28Independently at each occurrence, is selected from hydrogen, halogen, C1-C6 alkyl, C1-C3 haloalkyl, -OR5-CN and by-OH, -OR5or-NHR9Substituted C1-C6 alkyl;
R5、R9and R10As defined in claim 1;
R7is any one of the following group E;
Figure FDA0002830360930000171
wherein R is8And R14–R20As defined in claim 1.
7. The compound of any one of claims 1-5, having general formula VII
Figure FDA0002830360930000172
X, L therein1、R2、R6、R22、R23、R24And R25As defined in claim 1;
m、n、Y1、Y2、L2、R29、R30and Q1As defined in claim 3.
8. The compound according to any one of claims 1 and 2, having general formula VIII
Figure FDA0002830360930000181
X, L therein1、R2、R6And R10As defined in claim 1.
9. The compound of any one of claims 1-5, having general formula IX
Figure FDA0002830360930000182
Wherein L is1、R2、R6、R22、R23、R24And R25As defined in claim 1, and X3As defined in claim 6;
wherein Q2Is any one of the following group F;
Figure FDA0002830360930000183
R31and R32Absent OR independently at each occurrence selected from hydrogen, -OR5Halogen, -N (R)5)R5、-NR9R12、-NH(C=O)R5、-(C=O)NH2Aryl, heteroaryl, heterocyclyl, C1-C6 alkyl and substituted with-OH or-NH2Substituted C1-C6 alkyl;
wherein R is5、R9And R12As defined in claim 1.
10. The compound of any one of the preceding claims, having a structure selected from structures 1-279 as defined in table 11.
11. A pharmaceutical composition comprising a compound according to any one of claims 1-10 as active ingredient, together with at least one pharmaceutically acceptable carrier, excipient and/or diluent.
12. A compound according to claims 1-10 for use as a pharmaceutically active agent, wherein the pharmaceutically active agent preferably has inhibitory activity on cyclin dependent kinase 7(CDK 7).
13. Compound according to any one of claims 1 to 10 for use in a method for the prevention and/or treatment of a disease associated with the inhibition of apoptosis, aberrant transcriptional activity and/or cell cycle arrest caused by aberrant activity and/or overexpression of one or several Cyclin Dependent Kinases (CDKs), in particular cyclin dependent kinase 7(CDK7), wherein said disease is selected from the group consisting of proliferative diseases, infectious diseases including opportunistic diseases, immune diseases, autoimmune diseases and inflammatory diseases.
14. A compound for use according to claim 13, wherein the proliferative disease is a cancer, preferably a cancer selected from the group consisting of: adenocarcinoma, choroidal melanoma, acute leukemia, acoustic neuroma, ampulla, anal, astrocytoma, basal cell carcinoma, pancreatic, desmoid, bladder, bronchial, estrogen-dependent and independent breast cancer, burkitt's lymphoma, uterine body cancer, cancer of unknown primary tumor (CUP syndrome), colorectal cancer, small intestine, ovarian, endometrial, ependymoma, epithelial cancer types, ewing's tumor, gastrointestinal tumor, gastric cancer, gall bladder epithelial cancer, uterine cancer, cervical cancer, cervix, glioblastoma, gynecological tumor, otolaryngological tumor, hematological tumor, hairy cell leukemia, urinary tract cancer, skin testicular cancer, brain tumor (glioma), brain metastases, testicular cancer, pituitary tumor, carcinoid tumor, kaposi's sarcoma, laryngeal cancer, germ cell tumor, bone cancer, colorectal cancer, head and neck tumors (tumors of the ear, nose and throat region), colon cancer, craniopharyngeal tubular tumors, oral cavity cancers (cancers of the oral region and lips), cancers of the central nervous system, liver cancer, liver metastases, leukemia, eyelid tumors, lung cancer, lymphoma, stomach cancer, malignant melanoma, malignant neoplasias, gastrointestinal malignancies, breast cancer, rectal cancer, medulloblastoma, melanoma, meningioma, hodgkin/non-hodgkin lymphoma, mycosis fungoides, nasal cancer, schwannoma, neuroblastoma, kidney cancer, renal cell carcinoma, oligodendroglioma, esophageal cancer, osteolytic and osteogenic cancers, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, plasmacytoma, prostate cancer, pharyngeal cancer, rectal cancer, retinoblastoma, vaginal cancer, thyroid cancer, esophageal cancer, T-cell lymphoma, thymoma, ductal carcinoma, ocular tumor, urethral carcinoma, urological tumor, urothelial carcinoma, vulvar carcinoma, wart appearance, soft tissue tumor, soft tissue sarcoma, wilms tumor, cervical cancer, tongue cancer, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, lobular carcinoma in situ, small cell lung carcinoma, non-small cell lung carcinoma, bronchial adenoma, pleuropulmonary blastoma, mesothelioma, brain stem glioma, hypothalamic glioma, cerebellar astrocytoma, cerebral astrocytoma, neuroectodermal tumor, pineal tumor, uterine sarcoma, salivary gland carcinoma, anal gland adenocarcinoma, mast cell tumor, pelvic tumor, ureteral tumor, hereditary papillary renal carcinoma, sporadic renal carcinoma, intraocular melanoma, hepatocellular carcinoma, bile duct carcinoma, mixed hepatocyte cholangiocarcinoma, squamous cell carcinoma, malignant melanoma, merkel cell skin carcinoma, non-melanoma skin cancer, hypopharynx cancer, nasopharyngeal cancer, oropharyngeal cancer, oral cancer, squamous cell cancer, oral melanoma, AIDS-related lymphoma, cutaneous T-cell lymphoma, central nervous system lymphoma, malignant fibrous histiocytoma, lymphosarcoma, rhabdomyosarcoma, malignant histiocytosis, fibrosarcoma, angiosarcoma, hemangiopericyte tumor, Leiomyosarcoma (LMS), canine breast cancer, and feline breast cancer.
15. The compound for use according to claim 13, wherein the infectious disease, including opportunistic diseases, is selected from AIDS, adenovirus infection, alveolar echinococcosis (AHD), amebiasis, strongyloides angiostrongylosis, anisakiasis, anthrax, babesiosis, venocytosis, ascariasis, schistosomiasis (schistosomiasis), infections with human blastocysts, lyme borreliosis, botulism, braynard diarrhoea, brucellosis, Bovine Spongiform Encephalopathy (BSE), candidiasis, nematodiasis, Chronic Fatigue Syndrome (CFS), chagas disease, chicken pox, chlamydia pneumoniae infection, cholera, chronic fatigue syndrome, creutzfeldt-jakob disease (CJD), bronchoschistosomiasis, cutaneous larval migration disease (CLM), coccidioidomycosis, conjunctivitis, coxsackie virus a16(Cox a16), cryptococcosis, cryptosporidiosis, sporosis, and combinations thereof, West nile fever, circosiasis, encystosis, cytomegalovirus infection, dengue fever, dog-reptilation taenia infection, Ebola Hemorrhagic Fever (EHF), vesicular echinococcosis (AE), encephalitis, intracolonic amoeba infection, dispar amoeba infection, hamaun amoeba infection, bordetella infection, enterobiasis infection, enterovirus infection (polio/non-polio), Epstein Barr virus infection, escherichia coli infection, food-borne infection, foot and mouth disease, fungal dermatitis, fungal infection, gastroenteritis, group a streptococcosis, group B streptococcosis, hansenosis (leprosy disease), hantavirus lung syndrome, head lice infestation (pediculosis), helicobacter pylori infection, hematologic disease, hendra virus infection, hepatitis (HCV, HBV), herpes zoster (shingles), HIV infection, human ehrlichiosis, enterobiasis, hepatitis, Human parainfluenza virus infection, influenza, isocystosis, lassa fever, leishmaniasis, Visceral Leishmaniasis (VL), malaria, marburg hemorrhagic fever, measles, meningitis, Mycobacterium Avium Complex (MAC) infection, naesli infection, nosocomial infection, nonpathogenic intestinal amoeba infection, onchocerciasis, postorchidrosis, papilloma virus infection, parvovirus infection, plague, pneumocystis pneumonia (PCP), polyoma virus infection, Q fever, rabies, Respiratory Syncytial Virus (RSV) infection, rheumatic fever, rift valley fever, rotavirus infection, ascaris infection, salmonellosis, scabies, shigellosis, herpes zoster, narcolepsy, streptococcus infection, taenia infection, tetanus, toxic shock syndrome, tuberculosis, duodenum, vibrio parahaemolyticus infection, vibrio parainfluenza virus infection, measles fever, meningitis, herpes, tuberculosis, and infections of the duodenum, Vibrio septicemia, viral hemorrhagic fever, warts, water-borne infectious diseases, varicella-zoster virus infection, pertussis, and yellow fever.
16. A compound for use according to claim 13, wherein the immune and/or autoimmune disease is selected from: asthma, diabetes, rheumatic diseases, AIDS, rejection of transplanted organs and tissues, rhinitis, chronic obstructive pulmonary disease, osteoporosis, ulcerative colitis, sinusitis, lupus erythematosus, recurrent infections, atopic dermatitis/eczema and occupational allergies, food allergies, drug allergies, severe allergic reactions, allergies, manifestations of allergic disease, primary immunodeficiency, antibody deficient states, cell-mediated immunodeficiency, severe combined immunodeficiency, DiGeorge's syndrome, hyper IgE syndrome (HIES), Wiskott-Aldrich syndrome (WAS), ataxia telangiectasia, immune-mediated cancer, leukocyte deficiency, autoimmune diseases, Systemic Lupus Erythematosus (SLE), Rheumatoid Arthritis (RA), Multiple Sclerosis (MS), immune-mediated or type 1 diabetes, immune-mediated glomerulonephritis, scleroderma, pernicious anemia, alopecia, pemphigus vulgaris, myasthenia gravis, inflammatory bowel disease, crohn's disease, psoriasis, autoimmune thyroid disease, hashimoto's disease, dermatomyositis, goodpasture's syndrome (GPS), Myasthenia Gravis (MG), sympathetic ophthalmia, lens uveitis, chronic aggressive hepatitis, primary biliary cirrhosis, autoimmune hemolytic anemia, and weichhoff's disease.
17. The compound for use according to claim 13, wherein the inflammatory disease is caused, induced, caused and/or enhanced by bacteria, viruses, prions, parasites, fungi and/or is caused by a stimulatory, traumatic, metabolic, allergic, autoimmune or idiopathic factor.
18. The compound for use according to claim 13 or 17, wherein the inflammatory disease is selected from inflammatory diseases of the Central Nervous System (CNS), inflammatory rheumatic diseases, inflammatory diseases of the blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, uveitis inflammatory diseases and inflammatory diseases of the throat.
19. The compound for use according to any one of claims 13, 17 or 18, wherein the inflammatory disease is selected from inflammatory diseases of the Central Nervous System (CNS), inflammatory rheumatic diseases, inflammatory diseases of blood vessels, inflammatory diseases of the middle ear, inflammatory bowel diseases, inflammatory diseases of the skin, uveitis inflammatory diseases, inflammatory diseases of the throat, wherein preferably the inflammatory diseases are selected from abscesses, acanthamoeba infections, acne vulgaris, mycoses, acute inflammatory skin diseases, acute laryngeal infections in adults, acute multifocal squamous epitheliosis, acute (thermal) injuries, acute retinal necrosis, acute purulent otitis media, algal disorders, allergic contact dermatitis, amyloidosis vascular edema, ankylosing spondylitis, aspergillosis, atopic dermatitis, pseudorabies, autoantibodies in vasculitis, bacterial disorders, bacterial diseases, Bacterial laryngitis, bacterial meningitis, Behcet's Disease (BD), Mariothis armigera-like choroideremia, Creutzfeldt-Jakob disease, Borna disease, Brucella disease, bullous myringitis, bursitis, candidiasis, Canine fever encephalomyelitis in immature animals, Canine hemorrhagic fever, Canine herpes virus encephalomyelitis, cholesteatoma, Chronic Granulomatosis (CGD), chronic inflammatory skin disease, chronic relapsing encephalomyelitis, Chronic pyogenic otitis media, cicatricial pemphigoid (OCP), common upper respiratory tract infections, granuloma, Crohn's disease, cryptococcal disease, dermatomyositis, diphtheria, Discoid Lupus Erythematosus (DLE), drug-induced vasculitis, drug or hypersensitivity responses, encephalopathy, eosinophilic meningoencephalitis, polymorphic Erythrosis (EM), feline leukemia virus, feline immunodeficiency virus (IMV), Feline infectious peritonitis, feline poliovirus encephalitis, feline spongiform encephalopathy, fibromyalgia, Fuchs heterochroic uveitis, gastroesophageal (laryngopharyngeal) reflux disease, giant cell arteritis, melioidosis, glaucomatous ciliary syndrome, gonococcal granulomatosis, Granulomatous Meningoencephalitis (GME), herpes simplex, histoplasmosis, idiopathic diseases, idiopathic inflammatory disorders, immune and idiopathic disorders, infection of an immunocompromised host, infectious canine hepatitis, inhaled laryngitis, interstitial nephritis, irritant contact dermatitis, juvenile rheumatoid arthritis, kawasaki disease, larksrate virus encephalitis, laryngeal abscess, laryngotracheobronchitis, leishmaniasis, lens-induced uveitis, leprosy, leptospirosis, leukemia, lichen planus, lupus, lymphoma, meningitis, meningoencephalitis in rhotes, meningoencephalitis in dogs, lichen purpurea, meningitis, and meningitis, Miscellaneous meningitis/meningoencephalitis, microscopic polyangiitis, multifocal choroiditis, multifocal distemper encephalomyelitis in mature animals, multiple sclerosis, myotonic-dysphonia (MTD), fungal (fungal) disease, fungal disease of the CNS, necrotizing encephalitis, neosporosis, geriatric encephalitis, onchocerciasis, parasitic encephalomyelitis, parasitic infection, pars plana, parvoviral encephalitis, pediatric laryngitis, pollution and inhalation hypersensitivity, polymyositis, post-vaccine canine distemper encephalitis, prion protein-induced diseases, protozoon disease, protozoal encephalitis-encephalomyelitis, psoriasis, psoriatic arthritis, hamaun encephalitis, radiation injury, radiation laryngitis, radionecrosis, recurrent polychondritis, reiter syndrome, retinitis pigmentosa, retinoblastoma, rheumatoid arthritis, multiple sclerosis in mature animals, multiple sclerosis, myotonic dysfunction (MTD), multiple sclerosis, rickettsial disorder, rocky mountain spotted fever, Salmon Poisoning (SPD), sarcoidosis, schistosomiasis, scleroderma, rhinoscleroderma, creeping choroiditis, canine tremor, sjogren's syndrome, spastic laryngitis, spirochetal (syphilis) disease, spongiform dermatitis, sporotrichosis, steroid responsive meningitis-arteritis, Stevens-Johnson syndrome (SJS, severe EM), epiglottitis, sympathetic ophthalmia, paragonitis, syphilis, systemic vasculitis in sarcoidosis, takayasu arteritis, myositis (tenosynovitis), thromboangiitis obliterans (berger disease), tick-borne encephalitis in dogs, Toxic Epidermal Necrolysis (TEN), toxocarcinosis, toxoplasmosis, trauma, traumatic laryngitis, trichinosis, trypanosomiasis, tuberculosis, tularemia, ulcerative colitis, urticaria (urticaria), Vasculitis, vasculitis and malignancy, vasculitis and rheumatoid arthritis, vasculitis in idiopathic inflammatory myopathies, vasculitis of the central nervous system, vasculitis secondary to bacterial, fungal and parasitic infections, viral disorders, viral laryngitis, vitiligo, vocal cord abuse, vocal cord bleeding, Vogt-Koyanagi-Harada syndrome (VKH), Wegener's granulomatosis, and Whipple's disease.
CN201980039582.5A 2018-04-11 2019-04-11 Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases Pending CN112313235A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862656070P 2018-04-11 2018-04-11
US62/656,070 2018-04-11
PCT/EP2019/059302 WO2019197549A1 (en) 2018-04-11 2019-04-11 Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitor of cyclin dependent kinase

Publications (1)

Publication Number Publication Date
CN112313235A true CN112313235A (en) 2021-02-02

Family

ID=66334385

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201980039582.5A Pending CN112313235A (en) 2018-04-11 2019-04-11 Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases

Country Status (13)

Country Link
US (1) US11945822B2 (en)
EP (1) EP3774810A1 (en)
JP (2) JP2021521120A (en)
KR (1) KR20200141487A (en)
CN (1) CN112313235A (en)
AU (1) AU2019250507B2 (en)
BR (1) BR112020020768A2 (en)
CA (1) CA3095880A1 (en)
IL (1) IL277770B2 (en)
MX (1) MX2020010675A (en)
PH (1) PH12020551639A1 (en)
SG (1) SG11202009724YA (en)
WO (1) WO2019197549A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114195710A (en) * 2021-12-20 2022-03-18 上海彩迩文生化科技有限公司 Preparation method of quinoline aminomethyl compound
CN117756747A (en) * 2023-12-21 2024-03-26 济宁医学院 Preparation method of N-phenyl nitrogen-containing polycyclic quaternary ammonium salt compound
WO2024175024A1 (en) * 2023-02-21 2024-08-29 杭州德睿智药科技有限公司 New fused heterocyclic compound as cdks inhibitor and use thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220144841A1 (en) * 2019-03-13 2022-05-12 The Translational Genomics Research Institute Trisubstituted pyrazolo [1,5-a] pyrimidine compounds as cdk7 inhibitors
TW202146416A (en) * 2019-12-11 2021-12-16 德商拜耳廠股份有限公司 Pyrazolotriazines
GB201918541D0 (en) 2019-12-16 2020-01-29 Carrick Therapeutics Ltd Therapeutic compounds and their use
AU2020404326A1 (en) 2019-12-20 2022-08-11 Evopoint Biosciences Co., Ltd. Heterocyclic compound, and pharmaceutical composition thereof, preparation method therefor, intermediate thereof and application thereof
CN115485030A (en) * 2020-03-06 2022-12-16 拜耳公司 Imidazotriazines acting on cancer through inhibition of CDK12
CN112079781B (en) * 2020-09-30 2022-11-22 苏州康润医药有限公司 Synthesis method of 5-bromo-1-methyl-1H-pyrazol-3-amine
WO2022117504A1 (en) * 2020-12-02 2022-06-09 Qurient Co., Ltd. Compounds having cyclin-dependent kinase(cdk)-inhibitory function
GB202108572D0 (en) * 2021-06-16 2021-07-28 Carrick Therapeutics Ltd Therapeutic compounds and their use
WO2024151112A1 (en) * 2023-01-11 2024-07-18 주식회사 티움바이오 Pyrazolopyrimidine derivative as cyclin-dependent kinase 9 inhibitor

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1735614A (en) * 2002-09-04 2006-02-15 先灵公司 Pyrazolopyrimidines as cyclin-dependent kinase inhibitors
CN1922185A (en) * 2004-02-25 2007-02-28 先灵公司 Pyrazolotriazines as kinase inhibitors
CN101321757A (en) * 2005-10-06 2008-12-10 先灵公司 Pyrazolopyrimidines as cyclin dependent kinase inhibitors for the treatment of cancer
JP2009007342A (en) * 2007-06-01 2009-01-15 Mitsubishi Tanabe Pharma Corp Medicinal composition
CN101541804A (en) * 2006-10-04 2009-09-23 先灵公司 Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
WO2013128029A1 (en) * 2012-03-01 2013-09-06 Lead Discovery Center Gmbh Pharmaceutically active pyrazolo-triazine derivatives
CN104350054A (en) * 2012-03-01 2015-02-11 利德探索中心有限公司 Pyrazolo - triazine derivatives as selective cyclin- dependent kinase inhinitors
CN107530329A (en) * 2015-03-09 2018-01-02 奥瑞基尼探索技术有限公司 Pyrazolo [1,5 a] [1,3,5] triazine and pyrazolo [1,5 a] pyrimidine derivatives as CDK inhibitor

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US615968A (en) 1898-12-13 Apparatus for treating ores
US8673924B2 (en) * 2002-09-04 2014-03-18 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as cyclin dependent kinase inhibitors
US7119200B2 (en) 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
GB201403093D0 (en) * 2014-02-21 2014-04-09 Cancer Rec Tech Ltd Therapeutic compounds and their use
EP3129371B1 (en) 2014-04-05 2020-07-29 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
WO2019144149A2 (en) * 2018-01-22 2019-07-25 Fred Hutchinson Cancer Research Center Treatment methods for pancreatic tumors associated with the worst prognosis

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1735614A (en) * 2002-09-04 2006-02-15 先灵公司 Pyrazolopyrimidines as cyclin-dependent kinase inhibitors
CN1880317A (en) * 2002-09-04 2006-12-20 先灵公司 Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
CN1922185A (en) * 2004-02-25 2007-02-28 先灵公司 Pyrazolotriazines as kinase inhibitors
CN101321757A (en) * 2005-10-06 2008-12-10 先灵公司 Pyrazolopyrimidines as cyclin dependent kinase inhibitors for the treatment of cancer
CN101541804A (en) * 2006-10-04 2009-09-23 先灵公司 Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
JP2009007342A (en) * 2007-06-01 2009-01-15 Mitsubishi Tanabe Pharma Corp Medicinal composition
WO2013128029A1 (en) * 2012-03-01 2013-09-06 Lead Discovery Center Gmbh Pharmaceutically active pyrazolo-triazine derivatives
CN104350054A (en) * 2012-03-01 2015-02-11 利德探索中心有限公司 Pyrazolo - triazine derivatives as selective cyclin- dependent kinase inhinitors
CN107530329A (en) * 2015-03-09 2018-01-02 奥瑞基尼探索技术有限公司 Pyrazolo [1,5 a] [1,3,5] triazine and pyrazolo [1,5 a] pyrimidine derivatives as CDK inhibitor

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114195710A (en) * 2021-12-20 2022-03-18 上海彩迩文生化科技有限公司 Preparation method of quinoline aminomethyl compound
WO2024175024A1 (en) * 2023-02-21 2024-08-29 杭州德睿智药科技有限公司 New fused heterocyclic compound as cdks inhibitor and use thereof
CN117756747A (en) * 2023-12-21 2024-03-26 济宁医学院 Preparation method of N-phenyl nitrogen-containing polycyclic quaternary ammonium salt compound

Also Published As

Publication number Publication date
US20210147427A1 (en) 2021-05-20
KR20200141487A (en) 2020-12-18
PH12020551639A1 (en) 2021-07-05
IL277770A (en) 2020-11-30
EP3774810A1 (en) 2021-02-17
SG11202009724YA (en) 2020-10-29
CA3095880A1 (en) 2019-10-17
JP2021521120A (en) 2021-08-26
JP2023138858A (en) 2023-10-02
IL277770B1 (en) 2023-09-01
WO2019197549A1 (en) 2019-10-17
AU2019250507B2 (en) 2023-09-28
US11945822B2 (en) 2024-04-02
IL277770B2 (en) 2024-01-01
MX2020010675A (en) 2020-10-28
AU2019250507A1 (en) 2020-10-22
BR112020020768A2 (en) 2021-01-19

Similar Documents

Publication Publication Date Title
CN112313235A (en) Pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives as selective inhibitors of cyclin dependent kinases
JP7406501B2 (en) Pharmaceutically active pyrazolo-triazine and/or pyrazolo-pyrimidine derivatives
JP2021138734A (en) Novel compounds
CA2792508C (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors
AU2013227024B2 (en) Novel piperidine compound or salt thereof
CN113061132B (en) Condensed ring lactam compound, preparation method and application
AU2007287319A1 (en) Pyrimidone compounds as GSK-3 inhibitors
KR20150136497A (en) Tricyclic heterocycles as bet protein inhibitors
EA035499B1 (en) Novel glutaminase inhibitors
TW201706266A (en) CCR2 modulators
JP6733050B2 (en) Heteroaryl carboxamide compounds as inhibitors of RIPK2
US20190263828A1 (en) Heteroaromatic compounds as vanin inhibitors
KR20210080461A (en) Substituted Indole and Indazole Compounds
CN112079839A (en) Lactam derivative, preparation method and medical application thereof
US20240343732A1 (en) Compounds for degradation of cyclin-dependent kinase 7(cdk7)
RU2809779C2 (en) Pyrazolotriazine and/or pyrazolopyrimidine derivatives as a selective cyclin-dependent kinase inhibitor
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
AU2019257385B2 (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors
Ohashi Discovery of Novel Hedgehog Signaling Inhibitor
大橋知洋 Discovery of Novel Hedgehog Signaling Inhibitor
JP2022541819A (en) Thienopyridinyl and thiazolopyridinyl compounds useful as IRAK4 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination